1
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
2
|
Petrelli B, Oztürk A, Pind M, Ayele H, Fainsod A, Hicks GG. Genetically programmed retinoic acid deficiency during gastrulation phenocopies most known developmental defects due to acute prenatal alcohol exposure in FASD. Front Cell Dev Biol 2023; 11:1208279. [PMID: 37397253 PMCID: PMC10311642 DOI: 10.3389/fcell.2023.1208279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) arises from maternal consumption of alcohol during pregnancy affecting 2%-5% of the Western population. In Xenopus laevis studies, we showed that alcohol exposure during early gastrulation reduces retinoic acid (RA) levels at this critical embryonic stage inducing craniofacial malformations associated with Fetal Alcohol Syndrome. A genetic mouse model that induces a transient RA deficiency in the node during gastrulation is described. These mice recapitulate the phenotypes characteristic of prenatal alcohol exposure (PAE) suggesting a molecular etiology for the craniofacial malformations seen in children with FASD. Gsc +/Cyp26A1 mouse embryos have a reduced RA domain and expression in the developing frontonasal prominence region and delayed HoxA1 and HoxB1 expression at E8.5. These embryos also show aberrant neurofilament expression during cranial nerve formation at E10.5 and have significant FASD sentinel-like craniofacial phenotypes at E18.5. Gsc +/Cyp26A1 mice develop severe maxillary malocclusions in adulthood. Phenocopying the PAE-induced developmental malformations with a genetic model inducing RA deficiency during early gastrulation strongly supports the alcohol/vitamin A competition model as a major molecular etiology for the neurodevelopmental defects and craniofacial malformations seen in children with FASD.
Collapse
Affiliation(s)
- B. Petrelli
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - A. Oztürk
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - M. Pind
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - H. Ayele
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - A. Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - G. G. Hicks
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
3
|
Ietto G, Iori V, Gritti M, Inversini D, Costantino A, Izunza Barba S, Jiang ZG, Carcano G, Dalla Gasperina D, Pettinato G. Multicellular Liver Organoids: Generation and Importance of Diverse Specialized Cellular Components. Cells 2023; 12:1429. [PMID: 37408262 PMCID: PMC10217024 DOI: 10.3390/cells12101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
Over 40,000 patients in the United States are estimated to suffer from end-stage liver disease and acute hepatic failure, for which liver transplantation is the only available therapy. Human primary hepatocytes (HPH) have not been employed as a therapeutic tool due to the difficulty in growing and expanding them in vitro, their sensitivity to cold temperatures, and tendency to dedifferentiate following two-dimensional culture. The differentiation of human-induced pluripotent stem cells (hiPSCs) into liver organoids (LO) has emerged as a potential alternative to orthotropic liver transplantation (OLT). However, several factors limit the efficiency of liver differentiation from hiPSCs, including a low proportion of differentiated cells capable of reaching a mature phenotype, the poor reproducibility of existing differentiation protocols, and insufficient long-term viability in vitro and in vivo. This review will analyze various methodologies being developed to improve hepatic differentiation from hiPSCs into liver organoids, paying particular attention to the use of endothelial cells as supportive cells for their further maturation. Here, we demonstrate why differentiated liver organoids can be used as a research tool for drug testing and disease modeling, or employed as a bridge for liver transplantation following liver failure.
Collapse
Affiliation(s)
- Giuseppe Ietto
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Valentina Iori
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Mattia Gritti
- Department of General Surgery, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Davide Inversini
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Angelita Costantino
- Department of Drug and Health Sciences, University of Catania, 95124 Catania, Italy;
| | - Sofia Izunza Barba
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Z. Gordon Jiang
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Giulio Carcano
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Daniela Dalla Gasperina
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
- Department of Infectious Diseases, ASST-Sette Laghi, 21100 Varese, Italy
| | - Giuseppe Pettinato
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
4
|
Cao L, Wang R, Liu G, Zhang Y, Thorne RF, Zhang XD, Li J, Xia Y, Guo L, Shao F, Gu H, Wu M. Glycolytic Pfkp acts as a Lin41 protein kinase to promote endodermal differentiation of embryonic stem cells. EMBO Rep 2023; 24:e55683. [PMID: 36660859 PMCID: PMC9986826 DOI: 10.15252/embr.202255683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
Unveiling the principles governing embryonic stem cell (ESC) differentiation into specific lineages is critical for understanding embryonic development and for stem cell applications in regenerative medicine. Here, we establish an intersection between LIF-Stat3 signaling that is essential for maintaining murine (m) ESCs pluripotency, and the glycolytic enzyme, the platelet isoform of phosphofructokinase (Pfkp). In the pluripotent state, Stat3 transcriptionally suppresses Pfkp in mESCs while manipulating the cells to lift this repression results in differentiation towards the ectodermal lineage. Pfkp exhibits substrate specificity changes to act as a protein kinase, catalyzing serine phosphorylation of the developmental regulator Lin41. Such phosphorylation stabilizes Lin41 by impeding its autoubiquitination and proteasomal degradation, permitting Lin41-mediated binding and destabilization of mRNAs encoding ectodermal specification markers to favor the expression of endodermal specification genes. This provides new insights into the wiring of pluripotency-differentiation circuitry where Pfkp plays a role in germ layer specification during mESC differentiation.
Collapse
Affiliation(s)
- Leixi Cao
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Ruijie Wang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Guangzhi Liu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Yuwei Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Rick Francis Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
- School of Biomedical Sciences & PharmacyUniversity of NewcastleNewcastleNSWAustralia
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
- School of Environmental & Life SciencesUniversity of NewcastleNewcastleNSWAustralia
| | - Jinming Li
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Yang Xia
- Department of Immunology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Lili Guo
- Department of Immunology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Fengmin Shao
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
| | - Hao Gu
- Department of Immunology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Mian Wu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical ScienceZhengzhou UniversityZhengzhouChina
- School of Clinical MedicineHenan UniversityZhengzhouChina
- CAS Centre for Excellence in Molecular Cell Sciencethe First Affiliated Hospital of University of Science and Technology of ChinaHefeiChina
| |
Collapse
|
5
|
Takeishi K, Collin de l'Hortet A, Wang Y, Handa K, Guzman-Lepe J, Matsubara K, Morita K, Jang S, Haep N, Florentino RM, Yuan F, Fukumitsu K, Tobita K, Sun W, Franks J, Delgado ER, Shapiro EM, Fraunhoffer NA, Duncan AW, Yagi H, Mashimo T, Fox IJ, Soto-Gutierrez A. Assembly and Function of a Bioengineered Human Liver for Transplantation Generated Solely from Induced Pluripotent Stem Cells. Cell Rep 2021; 31:107711. [PMID: 32492423 DOI: 10.1016/j.celrep.2020.107711] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/17/2019] [Accepted: 05/08/2020] [Indexed: 12/22/2022] Open
Abstract
The availability of an autologous transplantable auxiliary liver would dramatically affect the treatment of liver disease. Assembly and function in vivo of a bioengineered human liver derived from induced pluripotent stem cells (iPSCs) has not been previously described. By improving methods for liver decellularization, recellularization, and differentiation of different liver cellular lineages of human iPSCs in an organ-like environment, we generated functional engineered human mini livers and performed transplantation in a rat model. Whereas previous studies recellularized liver scaffolds largely with rodent hepatocytes, we repopulated not only the parenchyma with human iPSC-hepatocytes but also the vascular system with human iPS-endothelial cells, and the bile duct network with human iPSC-biliary epithelial cells. The regenerated human iPSC-derived mini liver containing multiple cell types was tested in vivo and remained functional for 4 days after auxiliary liver transplantation in immunocompromised, engineered (IL2rg-/-) rats.
Collapse
Affiliation(s)
- Kazuki Takeishi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | - Yang Wang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Kan Handa
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jorge Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kentaro Matsubara
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kazutoyo Morita
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sae Jang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nils Haep
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rodrigo M Florentino
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| | - Fangchao Yuan
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ken Fukumitsu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kimimasa Tobita
- Department of Bioengineering and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15201, USA
| | - Wendell Sun
- LifeCell Corporation, Branchburg, NJ 08876, USA
| | - Jonathan Franks
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA
| | - Evan R Delgado
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3110, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Erik M Shapiro
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Nicolas A Fraunhoffer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Facultad de Ciencias de la Salud, Carrera de Medicina, Universidad Maimónides, Ciudad Autónoma de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires 1001, Argentina
| | - Andrew W Duncan
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3110, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hiroshi Yagi
- Department of Surgery, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, University of Tokyo, Tokyo 158-8557, Japan
| | - Ira J Fox
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3110, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3110, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
6
|
Ullah I, Seo K, Wi H, Kim Y, Lee S, Ock SA. Induction of the differentiation of porcine bone marrow mesenchymal stem cells into premature hepatocyte-like cells in an indirect coculture system with primary hepatocytes. Anim Cells Syst (Seoul) 2020; 24:289-298. [PMID: 33209203 PMCID: PMC7646558 DOI: 10.1080/19768354.2020.1823473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver transplantation is currently the only option for patients with end-stage liver disease. Thus, other alternate therapeutic strategies are needed. Bone marrow mesenchymal stem cells (BM-MSCs) are nonhematopoietic cells present in the bone marrow stroma that serve as precursors cells for various other cells. In this study, we evaluated the differentiation of porcine BM-MSCs into hepatocyte-like cells using three types of culture systems: hepatic induction medium (HIM), HIM/primary hepatocyte culture supernatant (HCS; 1:1 ratio), and a hepatocyte coculture system (HCCS; primary hepatocytes in the upper chamber, and BM-MSCs in the lower chamber). Primary hepatocytes were isolated from anesthetized healthy 1-month-old pigs by enzymatic digestion. Hepatic-specific marker expression (albumin [ALB], transferrin [TF], α-fetoprotein [AFP]), glycogen storage, low-density lipoprotein, and indocyanine green uptake were evaluated. Upregulation of hepatic-specific markers (ALB, TF, and AFP) was observed by real-time polymerase chain reaction in the HCCS group. Periodic acid-Schiff staining revealed enhanced glycogen storage in hepatocyte-like cells from the HCCS group compared with that from the HIM/HCS group. Furthermore, hepatocyte like-cells in the HCCS group showed improved LDL and ICG uptake than those in the other groups. Overall, our current study revealed that indirect coculture of primary hepatocytes and BM-MSCs enhanced the differentiation efficacy of BM-MSCs into hepatocyte-like cells by unknown useful soluble factors, including paracrine factors.
Collapse
Affiliation(s)
- Imran Ullah
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea.,Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Kangmin Seo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Hayeon Wi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Youngim Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Seunghoon Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| |
Collapse
|
7
|
Özkan A, Stolley D, Cressman ENK, McMillin M, DeMorrow S, Yankeelov TE, Rylander MN. The Influence of Chronic Liver Diseases on Hepatic Vasculature: A Liver-on-a-chip Review. MICROMACHINES 2020; 11:E487. [PMID: 32397454 PMCID: PMC7281532 DOI: 10.3390/mi11050487] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
In chronic liver diseases and hepatocellular carcinoma, the cells and extracellular matrix of the liver undergo significant alteration in response to chronic injury. Recent literature has highlighted the critical, but less studied, role of the liver vasculature in the progression of chronic liver diseases. Recent advancements in liver-on-a-chip systems has allowed in depth investigation of the role that the hepatic vasculature plays both in response to, and progression of, chronic liver disease. In this review, we first introduce the structure, gradients, mechanical properties, and cellular composition of the liver and describe how these factors influence the vasculature. We summarize state-of-the-art vascularized liver-on-a-chip platforms for investigating biological models of chronic liver disease and their influence on the liver sinusoidal endothelial cells of the hepatic vasculature. We conclude with a discussion of how future developments in the field may affect the study of chronic liver diseases, and drug development and testing.
Collapse
Affiliation(s)
- Alican Özkan
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Danielle Stolley
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Erik N K Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew McMillin
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Sharon DeMorrow
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
- Departments of Diagnostic Medicine, The University of Texas, Austin, TX 78712, USA
- Department of Oncology, The University of Texas, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, TX 78712, USA
| | - Marissa Nichole Rylander
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
| |
Collapse
|
8
|
Afshari A, Shamdani S, Uzan G, Naserian S, Azarpira N. Different approaches for transformation of mesenchymal stem cells into hepatocyte-like cells. Stem Cell Res Ther 2020; 11:54. [PMID: 32033595 PMCID: PMC7007672 DOI: 10.1186/s13287-020-1555-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/21/2019] [Accepted: 01/07/2020] [Indexed: 01/09/2023] Open
Abstract
Due to the prominent role of the liver in the body and detoxification, its functionality can be affected in an irreversible manner by diseases. This phenomenon renders the liver to stop working, leading to morbidity and mortality. Therefore, liver transplantation is the only way to tackle this issue.In order to compensate for the lack of adequate healthy liver tissue for transplantation, therapeutic approaches such as hepatocyte transplantation have been proposed as an alternative. Recognizing the fact that mesenchymal stem cells are adult stem cells with the capacity to differentiate into several cell types, different methods have been invented to produce hepatocyte-like cells from mesenchymal stem cells. They can be divided into three main categories, such as addition of cytokines and growth factors, genetic modifications, and adjustment of microenvironment as well as physical parameters.In this review, we attempted to introduce diverse efficient methods for differentiating mesenchymal stem cells and their capability for transformation into hepatocyte-like cells.
Collapse
Affiliation(s)
- Afsoon Afshari
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili street, Shiraz, Iran
| | - Sara Shamdani
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807, Villejuif, France.,SivanCell, Tehran, Iran.,CellMedEx, Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807, Villejuif, France
| | - Sina Naserian
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807, Villejuif, France.,SivanCell, Tehran, Iran.,CellMedEx, Saint Maur Des Fossés, France
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili street, Shiraz, Iran.
| |
Collapse
|
9
|
Mirdamadi ES, Kalhori D, Zakeri N, Azarpira N, Solati-Hashjin M. Liver Tissue Engineering as an Emerging Alternative for Liver Disease Treatment. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:145-163. [PMID: 31797731 DOI: 10.1089/ten.teb.2019.0233] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic liver diseases affect thousands of lives throughout the world every year. The shortage of liver donors for transplantation has been the main driving force to employ alternative methods such as liver tissue engineering (LTE) in fabricating a three-dimensional transplantable liver tissue or enhancing cell delivery techniques alleviating the need for liver donors. LTE consists of three components, cells, ECM (extracellular matrix), and signaling molecules, which we discuss the first and second. The three most common cell sources used in LTE are human and animal primary hepatocytes, and stem cells for different applications. Two major categories of ECM are used to mimic the microenvironment of these cells, named scaffolds and microbeads. Scaffolds have been made by numerous methods with a wide range of synthetic and natural biomaterials. Cell encapsulation has also been utilized by many polymeric biomaterials. To investigate their functions, many properties have been discussed in the literature, such as biochemical, geometrical, and mechanical properties, in both of these categories. Overall, LTE shows excellent potential in assisting hepatic disorders. However, some challenges exist that prevent the practical use of it clinically, making LTE an ongoing research subject in the scientific society.
Collapse
Affiliation(s)
- 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
| | - Nima Zakeri
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
10
|
Naskar S, Kumaran V, Markandeya YS, Mehta B, Basu B. Neurogenesis-on-Chip: Electric field modulated transdifferentiation of human mesenchymal stem cell and mouse muscle precursor cell coculture. Biomaterials 2019; 226:119522. [PMID: 31669894 DOI: 10.1016/j.biomaterials.2019.119522] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
Abstract
A number of bioengineering strategies, using biophysical stimulation, are being explored to guide the human mesenchymal stem cells (hMScs) into different lineages. In this context, we have limited understanding on the transdifferentiation of matured cells to another functional-cell type, when grown with stem cells, in a constrained cellular microenvironment under biophysical stimulation. While addressing such aspects, the present work reports the influence of the electric field (EF) stimulation on the phenotypic and functionality modulation of the coculture of murine myoblasts (C2C12) with hMScs [hMSc:C2C12=1:10] in a custom designed polymethylmethacrylate (PMMA) based microfluidic device with in-built metal electrodes. The quantitative and qualitative analysis of the immunofluorescence study confirms that the cocultured cells in the conditioned medium with astrocytic feed, exhibit differentiation towards neural-committed cells under biophysical stimulation in the range of the endogenous physiological electric field strength (8 ± 0.06 mV/mm). The control experiments using similar culture protocols revealed that while C2C12 monoculture exhibited myotube-like fused structures, the hMScs exhibited the neurosphere-like clusters with SOX2, nestin, βIII-tubulin expression. The electrophysiological study indicates the significant role of intercellular calcium signalling among the differentiated cells towards transdifferentiation. Furthermore, the depolarization induced calcium influx strongly supports neural-like behaviour for the electric field stimulated cells in coculture. The intriguing results are explained in terms of the paracrine signalling among the transdifferentiated cells in the electric field stimulated cellular microenvironment. In summary, the present study establishes the potential for neurogenesis on-chip for the coculture of hMSc and C2C12 cells under tailored electric field stimulation, in vitro.
Collapse
Affiliation(s)
- Sharmistha Naskar
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India; Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India; Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, 560012, India; Centres of Excellence and Innovation in Biotechnology - Translational Centre on Biomaterials for Orthopaedic and Dental Applications, Materials Research Centre, IISc, Bangalore, India
| | - Viswanathan Kumaran
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Yogananda S Markandeya
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India
| | - Bikramjit Basu
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India; Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, 560012, India; Centres of Excellence and Innovation in Biotechnology - Translational Centre on Biomaterials for Orthopaedic and Dental Applications, Materials Research Centre, IISc, Bangalore, India.
| |
Collapse
|
11
|
Torresi J, Tran BM, Christiansen D, Earnest-Silveira L, Schwab RHM, Vincan E. HBV-related hepatocarcinogenesis: the role of signalling pathways and innovative ex vivo research models. BMC Cancer 2019; 19:707. [PMID: 31319796 PMCID: PMC6637598 DOI: 10.1186/s12885-019-5916-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is the leading cause of liver cancer, but the mechanisms by which HBV causes liver cancer are poorly understood and chemotherapeutic strategies to cure liver cancer are not available. A better understanding of how HBV requisitions cellular components in the liver will identify novel therapeutic targets for HBV associated hepatocellular carcinoma (HCC). MAIN BODY The development of HCC involves deregulation in several cellular signalling pathways including Wnt/FZD/β-catenin, PI3K/Akt/mTOR, IRS1/IGF, and Ras/Raf/MAPK. HBV is known to dysregulate several hepatocyte pathways and cell cycle regulation resulting in HCC development. A number of these HBV induced changes are also mediated through the Wnt/FZD/β-catenin pathway. The lack of a suitable human liver model for the study of HBV has hampered research into understanding pathogenesis of HBV. Primary human hepatocytes provide one option; however, these cells are prone to losing their hepatic functionality and their ability to support HBV replication. Another approach involves induced-pluripotent stem (iPS) cell-derived hepatocytes. However, iPS technology relies on retroviruses or lentiviruses for effective gene delivery and pose the risk of activating a range of oncogenes. Liver organoids developed from patient-derived liver tissues provide a significant advance in HCC research. Liver organoids retain the characteristics of their original tissue, undergo unlimited expansion, can be differentiated into mature hepatocytes and are susceptible to natural infection with HBV. CONCLUSION By utilizing new ex vivo techniques like liver organoids it will become possible to develop improved and personalized therapeutic approaches that will improve HCC outcomes and potentially lead to a cure for HBV.
Collapse
Affiliation(s)
- Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Bang Manh Tran
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Dale Christiansen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Linda Earnest-Silveira
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Renate Hilda Marianne Schwab
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Elizabeth Vincan
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010 Australia
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6845 Australia
| |
Collapse
|
12
|
Agarwal N, Popovic B, Martucci NJ, Fraunhoffer NA, Soto-Gutierrez A. Biofabrication of Autologous Human Hepatocytes for Transplantation: How Do We Get There? Gene Expr 2019; 19:89-95. [PMID: 30143060 PMCID: PMC6466180 DOI: 10.3727/105221618x15350366478989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Directed differentiation of hepatocytes from induced pluripotent stem cells (iPSCs) holds promise as source material for treating some liver disorders. The unlimited availability of perfectly differentiated iPSC-derived hepatocytes will dramatically facilitate cell therapies. While systems to manufacture large quantities of iPSC-derived cells have been developed, we have been unable to generate and maintain stable and mature adult liver cells ex vivo. This short review highlights important challenges and possible solutions to the current state of hepatocyte biofabrication for cellular therapies to treat liver diseases. Successful cell transplantation will require optimizing the best cell function, overcoming limitations to cell numbers and safety, as well as a number of other challenges. Collaboration among scientists, clinicians, and industry is critical for generating new autologous stem cell-based therapies to treat liver diseases.
Collapse
Affiliation(s)
- Nandini Agarwal
- *School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
- †Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Branimir Popovic
- †Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nicole J. Martucci
- †Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nicolas A. Fraunhoffer
- †Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- ‡Facultad de Ciencias de la Salud, Carrera de Medicina, Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- §Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | | |
Collapse
|
13
|
Raasch M, Fritsche E, Kurtz A, Bauer M, Mosig AS. Microphysiological systems meet hiPSC technology - New tools for disease modeling of liver infections in basic research and drug development. Adv Drug Deliv Rev 2019; 140:51-67. [PMID: 29908880 DOI: 10.1016/j.addr.2018.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
Complex cell culture models such as microphysiological models (MPS) mimicking human liver functionality in vitro are in the spotlight as alternative to conventional cell culture and animal models. Promising techniques like microfluidic cell culture or micropatterning by 3D bioprinting are gaining increasing importance for the development of MPS to address the needs for more predictivity and cost efficiency. In this context, human induced pluripotent stem cells (hiPSCs) offer new perspectives for the development of advanced liver-on-chip systems by recreating an in vivo like microenvironment that supports the reliable differentiation of hiPSCs to hepatocyte-like cells (HLC). In this review we will summarize current protocols of HLC generation and highlight recently established MPS suitable to resemble physiological hepatocyte function in vitro. In addition, we are discussing potential applications of liver MPS for disease modeling related to systemic or direct liver infections and the use of MPS in testing of new drug candidates.
Collapse
|
14
|
Bandi S, Tchaikovskaya T, Gupta S. Hepatic differentiation of human pluripotent stem cells by developmental stage-related metabolomics products. Differentiation 2019; 105:54-70. [PMID: 30776728 DOI: 10.1016/j.diff.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
Endogenous cell signals regulate tissue homeostasis and are significant for directing the fate of stem cells. During liver development, cytokines released from various cell types are critical for stem/progenitor cell differentiation and lineage expansions. To determine mechanisms in these stage-specific lineage interactions, we modeled potential effects of soluble signals derived from immortalized human fetal liver parenchymal cells on stem cells, including embryonic and induced pluripotent stem cells. For identifying lineage conversion and maturation, we utilized conventional assays of cell morphology, gene expression analysis and lineage markers. Molecular pathway analysis used functional genomics approaches. Metabolic properties were analyzed to determine the extent of hepatic differentiation. Cell transplantation studies were performed in mice with drug-induced acute liver failure to elicit benefits in hepatic support and tissue regeneration. These studies showed signals emanating from fetal liver cells induced hepatic differentiation in stem cells. Gene expression profiling and comparison of regulatory networks in immature and mature hepatocytes revealed stem cell-derived hepatocytes represented early fetal-like stage. Unexpectedly, differentiation-inducing soluble signals constituted metabolomics products and not proteins. In stem cells exposed to signals from fetal cells, mechanistic gene networks of upstream regulators decreased pluripotency, while simultaneously inducing mesenchymal and epithelial properties. The extent of metabolic and synthetic functions in stem cell-derived hepatocytes was sufficient for providing hepatic support along with promotion of tissue repair to rescue mice in acute liver failure. During this rescue, paracrine factors from transplanted cells contributed in stimulating liver regeneration. We concluded that hepatic differentiation of pluripotent stem cells with metabolomics products will be significant for developing therapies. The differentiation mechanisms involving metabolomics products could have an impact on advancing recruitment of stem/progenitor cells during tissue homeostasis.
Collapse
Affiliation(s)
- Sriram Bandi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Tatyana Tchaikovskaya
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Diabetes Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Irwin S. and Sylvia Chanin Institute for Cancer Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
15
|
Nahar S, Nakashima Y, Miyagi-Shiohira C, Kinjo T, Toyoda Z, Kobayashi N, Saitoh I, Watanabe M, Noguchi H, Fujita J. Cytokines in adipose-derived mesenchymal stem cells promote the healing of liver disease. World J Stem Cells 2018; 10:146-159. [PMID: 30631390 PMCID: PMC6325075 DOI: 10.4252/wjsc.v10.i11.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are a treatment cell source for patients with chronic liver injury. ADSCs are characterized by being harvested from the patient's own subcutaneous adipose tissue, a high cell yield (i.e., reduced immune rejection response), accumulation at a disease nidus, suppression of excessive immune response, production of various growth factors and cytokines, angiogenic effects, anti-apoptotic effects, and control of immune cells via cell-cell interaction. We previously showed that conditioned medium of ADSCs promoted hepatocyte proliferation and improved the liver function in a mouse model of acute liver failure. Furthermore, as found by many other groups, the administration of ADSCs improved liver tissue fibrosis in a mouse model of liver cirrhosis. A comprehensive protein expression analysis by liquid chromatography with tandem mass spectrometry showed that the various cytokines and chemokines produced by ADSCs promote the healing of liver disease. In this review, we examine the ability of expressed protein components of ADSCs to promote healing in cell therapy for liver disease. Previous studies demonstrated that ADSCs are a treatment cell source for patients with chronic liver injury. This review describes the various cytokines and chemokines produced by ADSCs that promote the healing of liver disease.
Collapse
Affiliation(s)
- Saifun Nahar
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Zensei Toyoda
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan
| | - Masami Watanabe
- Department of Urology, Okayama Univer sity Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| |
Collapse
|
16
|
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
|
17
|
Raju R, Chau D, Notelaers T, Myers CL, Verfaillie CM, Hu WS. In Vitro Pluripotent Stem Cell Differentiation to Hepatocyte Ceases Further Maturation at an Equivalent Stage of E15 in Mouse Embryonic Liver Development. Stem Cells Dev 2018; 27:910-921. [PMID: 29851366 DOI: 10.1089/scd.2017.0270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatocyte-like cells (HLCs) can be derived from pluripotent stem cells (PSCs) by sequential treatment of chemical cues to mimic the microenvironment of embryonic liver development. However, these HLCs do not reach the full maturity level of primary hepatocytes. In this study, we carried out a meta-analysis of cross-species transcriptome data of in vitro differentiation of human PSCs to HLCs and in vivo mouse embryonic liver development to identify the developmental stage at which HLC maturation was blocked at. Systematic variations were found associated with the data source and removed by batch correction. Using principal component analysis, HLCs from different stages of differentiation were aligned with mouse embryonic liver development chronologically. A "unified developmental time" (DT) scale was developed after aligning in vitro HLC differentiation and in vivo embryonic liver development. HLCs were found to cease further maturation at an equivalent stage of mouse embryonic day (E)13-15. Genes with discordant time dynamics were identified by aligning in vivo and in vitro data set onto a common DT scale. These genes may be targets of genetic intervention for enhancing the maturity of PSC-derived HLCs.
Collapse
Affiliation(s)
- Ravali Raju
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David Chau
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota.,3 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Tineke Notelaers
- 4 Department of Development and Regeneration, KU Leuven , Leuven, Belgium .,5 Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Chad L Myers
- 6 Department of Computer Science and Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Catherine M Verfaillie
- 4 Department of Development and Regeneration, KU Leuven , Leuven, Belgium .,5 Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Wei-Shou Hu
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
18
|
Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A. Liver 'organ on a chip'. Exp Cell Res 2018; 363:15-25. [PMID: 29291400 PMCID: PMC5944300 DOI: 10.1016/j.yexcr.2017.12.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems.
Collapse
Affiliation(s)
- Colin H Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
19
|
Chen J, Tschudy-Seney B, Ma X, Zern MA, Liu P, Duan Y. Salvianolic Acid B Enhances Hepatic Differentiation of Human Embryonic Stem Cells Through Upregulation of WNT Pathway and Inhibition of Notch Pathway. Stem Cells Dev 2018; 27:252-261. [PMID: 29256825 DOI: 10.1089/scd.2017.0168] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocytes differentiated from human embryonic stem cells (ESCs) could provide a powerful tool for enabling cell-based therapies, studying the mechanisms underlying human liver development and disease, and testing the efficacy and safety of pharmaceuticals. However, currently most in vitro protocols yield hepatocytes with low levels of liver function. In this study, we investigated the potential of Salvianolic acid B (Sal B), an active pharmaceutical compound present in Salvia miltiorrhiza, which has been shown to have an antifibrotic effect in previous studies, to enhance hepatocyte differentiation from human ESCs. After treatment with Sal B, albumin expression and secretion were consistently increased, indicating that Sal B could promote hepatocyte differentiation process. Expression of a large number of important phase 1 and 2 metabolizing enzymes and phase 3 transporters was also increased in treated cells, indicating an enhanced biotransformation function. Our investigations further revealed the activation of Wnt pathway in treated cells, as determined by upregulation of Wnts, which increased amounts of nuclear β-catenin. This increased nuclear β-catenin led in turn to the enhanced expression of T cell factor (TCF) 3 and lymphoid enhancer-binding factor (LEF) 1 which upregulated their downstream targets, cyclin D1 and c-Myc. Notch receptors (Notch1, Notch3), Notch ligand (Jagged2), and Notch receptor targets [hairy and enhancer of split (Hes) 1, 5] were downregulated in treated cells, suggesting that Notch pathway was inhibited. Consistent with the inhibition of Notch pathway, expression of cholangiocyte marker, CK7, was significantly reduced by treatment with Sal B. Numb, a direct transcriptional target of Wnt pathway and a negative regulator of Notch pathway, was upregulated, consistent with activation of Wnt signaling and suppression of Notch signaling. In conclusion, our study demonstrated that Sal B enhanced hepatocyte differentiation from human ESCs through activation of Wnt pathway and inhibition of Notch pathway. Therefore, this study suggests that Sal B can be used as a potential agent to generate more mature hepatocytes for cell-based therapeutics and pharmaceutical studies.
Collapse
Affiliation(s)
- Jiamei Chen
- 1 Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine , Shanghai, China .,2 Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China .,3 Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China .,4 E-institutes of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine , Shanghai, China .,5 Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Benjamin Tschudy-Seney
- 5 Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Xiaocui Ma
- 5 Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Mark A Zern
- 5 Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Ping Liu
- 1 Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine , Shanghai, China .,2 Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China .,3 Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai, China .,4 E-institutes of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Yuyou Duan
- 5 Department of Internal Medicine, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,6 Department of Dermatology, Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| |
Collapse
|
20
|
Human embryoid bodies to hepatocyte-like clusters: Preparing for translation. LIVER RESEARCH 2017. [DOI: 10.1016/j.livres.2017.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
21
|
Khazali AS, Clark AM, Wells A. A Pathway to Personalizing Therapy for Metastases Using Liver-on-a-Chip Platforms. Stem Cell Rev Rep 2017; 13:364-380. [PMID: 28425064 PMCID: PMC5484059 DOI: 10.1007/s12015-017-9735-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metastasis accounts for most cancer-related deaths. The majority of solid cancers, including those of the breast, colorectum, prostate and skin, metastasize at significant levels to the liver due to its hemodynamic as well as tumor permissive microenvironmental properties. As this occurs prior to detection and treatment of the primary tumor, we need to target liver metastases to improve patients' outcomes. Animal models, while proven to be useful in mechanistic studies, do not represent the heterogeneity of human population especially in drug metabolism lack proper human cell-cell interactions, and this gap between animals and humans results in costly and inefficient drug discovery. This underscores the need to accurately model the human liver for disease studies and drug development. Further, the occurrence of liver metastases is influenced by the primary tumor type, sex and race; thus, modeling these specific settings will facilitate the development of personalized/targeted medicine for each specific group. We have adapted such all-human 3D ex vivo hepatic microphysiological system (MPS) (a.k.a. liver-on-a-chip) to investigate human micrometastases. This review focuses on the sources of liver resident cells, especially the iPS cell-derived hepatocytes, and examines some of the advantages and disadvantages of these sources. In addition, this review also examines other potential challenges and limitations in modeling human liver.
Collapse
Affiliation(s)
- A S Khazali
- Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace St, Pittsburgh, PA, 15261, USA
| | - A M Clark
- Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace St, Pittsburgh, PA, 15261, USA
| | - A Wells
- Department of Pathology, University of Pittsburgh, S711 Scaife Hall, 3550 Terrace St, Pittsburgh, PA, 15261, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
- Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Perez RA, Jung CR, Kim HW. Biomaterials and Culture Technologies for Regenerative Therapy of Liver Tissue. Adv Healthc Mater 2017; 6. [PMID: 27860372 DOI: 10.1002/adhm.201600791] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/10/2016] [Indexed: 12/18/2022]
Abstract
Regenerative approach has emerged to substitute the current extracorporeal technologies for the treatment of diseased and damaged liver tissue. This is based on the use of biomaterials that modulate the responses of hepatic cells through the unique matrix properties tuned to recapitulate regenerative functions. Cells in liver preserve their phenotype or differentiate through the interactions with extracellular matrix molecules. Therefore, the intrinsic properties of the engineered biomaterials, such as stiffness and surface topography, need to be tailored to induce appropriate cellular functions. The matrix physical stimuli can be combined with biochemical cues, such as immobilized functional groups or the delivered actions of signaling molecules. Furthermore, the external modulation of cells, through cocultures with nonparenchymal cells (e.g., endothelial cells) that can signal bioactive molecules, is another promising avenue to regenerate liver tissue. This review disseminates the recent approaches of regenerating liver tissue, with a focus on the development of biomaterials and the related culture technologies.
Collapse
Affiliation(s)
- Roman A. Perez
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Regenerative Medicine Research Institute; Universitat Internacional de Catalunya; Barcelona 08017 Spain
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
| | - Cho-Rok Jung
- Gene Therapy Research Unit; KRIBB; 125 Gwahak-ro Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
- Department of Biomaterials Science; Dankook University Dental College; Cheonan 330-714 Republic of Korea
| |
Collapse
|
23
|
Wang Y, Lee JH, Shirahama H, Seo J, Glenn JS, Cho NJ. Extracellular Matrix Functionalization and Huh-7.5 Cell Coculture Promote the Hepatic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells in a 3D ICC Hydrogel Scaffold. ACS Biomater Sci Eng 2016; 2:2255-2265. [PMID: 33465898 DOI: 10.1021/acsbiomaterials.6b00487] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this study, we constructed a microporous hydrogel scaffold with hexagonally packed interconnected cavities and extracellular matrix (ECM)-functionalized interior surface, and systematically investigated the hepatic differentiation of human adipose-derived mesenchymal stem cells (hAD-MSCs) under the influence of three key factors: three-dimensional (3D) geometry, ECM presence, and coculture with hepatocyte-derived cell line. Results confirmed that (i) hepatic differentiation of hAD-MSC is more efficient in a 3D microporous scaffold than in 2D monolayer culture; (ii) the presence of both ECM components (fibronectin and collagen-I) in the scaffold is superior to collagen-I only, highlighting the importance of fibronectin; and (iii) coculture with Huh-7.5 hepatocyte-derived cells promoted liver-specific functions of the hAD-MSC-derived hepatocytes. The optimized differentiation process only took 21 days to complete, a time length that is shorter or at least comparable to previous reports, and more importantly, yielded an albumin production more than 10-fold higher than conventional 2D culture. Our approach of optimizing hAD-MSC hepatic differentiation could provide a potential solution to the challenges such as hepatocyte transplantation or the establishment of human physiologically relevant liver models in vitro.
Collapse
Affiliation(s)
- Yan Wang
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore
| | - Jae-Ho Lee
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore
| | - Hitomi Shirahama
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore
| | - Jeongeun Seo
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore
| | - Jeffrey S Glenn
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Alway Building, Room M211, 300 Pasteur Drive, Stanford, California 94305, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Fairchild Building, D300, 299 Campus Drive, Stanford, California 94305, United States
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue 639798, Singapore.,School of Chemical and Biomolecular Engineering, Nanyang Technological University, 62 Nanyang Avenue 637459, Singapore
| |
Collapse
|
24
|
Zhang SB, Sun X, Wu Q, Wu JP, Chen HY. Impaired Capacity of Fibroblasts to Support Airway Epithelial Progenitors in Bronchiolitis Obliterans Syndrome. Chin Med J (Engl) 2016; 129:2040-4. [PMID: 27569228 PMCID: PMC5009585 DOI: 10.4103/0366-6999.189058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) often develops in transplant patients and results in injury to the respiratory and terminal airway epithelium. Owing to its rising incidence, the pathogenesis of BOS is currently an area of intensive research. Studies have shown that injury to the respiratory epithelium results in dysregulation of epithelial repair. Airway epithelial regeneration is supported by stromal cells, including fibroblasts. This study aimed to investigate whether the supportive role of lung fibroblasts is altered in BOS. METHODS Suspensions of lung cells were prepared by enzyme digestion. Lung progenitor cells (LPCs) were separated by fluorescence-activated cell sorting. Lung fibroblasts from patients with BOS or healthy controls were mixed with sorted mouse LPCs to compare the colony-forming efficiency of LPCs by counting the number of colonies with a diameter of ≥50 μm in each culture. Statistical analyses were performed using the SPSS 17.0 software (SPSS Inc., USA). The paired Student's t-test was used to test for statistical significance. RESULTS LPCs were isolated with the surface phenotype of CD31-CD34-CD45- EpCAM+Sca-1+. The colony-forming efficiency of LPCs was significantly reduced when co-cultured with fibroblasts isolated from patients with BOS. The addition of SB431542 increased the colony-forming efficiency of LPCs to 1.8%; however, it was still significantly less than that in co-culture with healthy control fibroblasts (P < 0.05). CONCLUSION The epithelial-supportive capacity of fibroblasts is impaired in the development of BOS and suggest that inefficient repair of airway epithelium could contribute to persistent airway inflammation in BOS.
Collapse
Affiliation(s)
- Su-Bei Zhang
- Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Xin Sun
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Qi Wu
- Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin 300350, China
- Address for correspondence: Prof. Qi Wu, Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin 300350, China E-Mail:
| | - Jun-Ping Wu
- Department of Respiratory, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Huai-Yong Chen
- Department of Basic Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| |
Collapse
|
25
|
Hu M, Li S, Menon S, Liu B, Hu MS, Longaker MT, Lorenz HP. Expansion and Hepatic Differentiation of Adult Blood-Derived CD34+ Progenitor Cells and Promotion of Liver Regeneration After Acute Injury. Stem Cells Transl Med 2016; 5:723-32. [PMID: 27075766 PMCID: PMC4878335 DOI: 10.5966/sctm.2015-0268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/13/2016] [Indexed: 12/27/2022] Open
Abstract
A new group of blood-derived CD34+ progenitor cells (BDPCs) with the ability to expand and differentiate into functional hepatocyte-like cells and promote liver regeneration is reported. With their ease of access, application through the peripheral blood, and the capability of rapid expansion and hepatic differentiation, BDPCs have great potential as a cell-based therapy for liver disease. The low availability of functional hepatocytes has been an unmet demand for basic scientific research, new drug development, and cell-based clinical applications for decades. Because of the inability to expand hepatocytes in vitro, alternative sources of hepatocytes are a focus of liver regenerative medicine. We report a new group of blood-derived CD34+ progenitor cells (BDPCs) that have the ability to expand and differentiate into functional hepatocyte-like cells and promote liver regeneration. BDPCs were obtained from the peripheral blood of an adult mouse with expression of surface markers CD34, CD45, Sca-1, c-kit, and Thy1.1. BDPCs can proliferate in vitro and differentiate into hepatocyte-like cells expressing hepatocyte markers, including CK8, CK18, CK19, α-fetoprotein, integrin-β1, and A6. The differentiated BDPCs (dBDPCs) also display liver-specific functional activities, such as glycogen storage, urea production, and albumin secretion. dBDPCs have cytochrome P450 activity and express specific hepatic transcription factors, such as hepatic nuclear factor 1α. To demonstrate liver regenerative activity, dBDPCs were injected into mice with severe acute liver damage caused by a high-dose injection of carbon tetrachloride (CCl4). dBDPC treatment rescued the mice from severe acute liver injury, increased survival, and induced liver regeneration. Because of their ease of access and application through peripheral blood and their capability of rapid expansion and hepatic differentiation, BDPCs have great potential as a cell-based therapy for liver disease. Significance Hematopoietic stem/progenitor cell expansion and tissue-specific differentiation in vitro are challenges in regenerative medicine, although stem cell therapy has raised hope for the treatment of liver diseases by overcoming the scarcity of hepatocytes. This study identified and characterized a group of blood-derived progenitor cells (BDPCs) from the peripheral blood of an adult mouse. The CD34+ progenitor-dominant BDPCs were rapidly expanded and hepatically differentiated into functional hepatocyte-like cells with our established coculture system. BDPC treatment increased animal survival and produced full regeneration in a severe liver injury mouse model caused by CCl4. BDPCs could have potential for liver cell therapies.
Collapse
Affiliation(s)
- Min Hu
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Shaowei Li
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Siddharth Menon
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Bo Liu
- Division of Pediatric Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Michael S Hu
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA Department of Surgery, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Michael T Longaker
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - H Peter Lorenz
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
26
|
Siltanen C, Yaghoobi M, Haque A, You J, Lowen J, Soleimani M, Revzin A. Microfluidic fabrication of bioactive microgels for rapid formation and enhanced differentiation of stem cell spheroids. Acta Biomater 2016; 34:125-132. [PMID: 26774761 DOI: 10.1016/j.actbio.2016.01.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/11/2015] [Accepted: 01/12/2016] [Indexed: 01/17/2023]
Abstract
A major challenge in tissue engineering is to develop robust protocols for differentiating ES and iPS cells to functional adult tissues at a clinically relevant scale. The goal of this study is to develop a high throughput platform for generating bioactive, stem cell-laden microgels to direct differentiation in a well-defined microenvironment. We describe a droplet microfluidics system for fabricating microgels composed of polyethylene glycol and heparin, with tunable geometric, mechanical, and chemical properties, at kHz rates. Heparin-containing hydrogel particles sequestered growth factors Nodal and FGF-2, which are implicated in specifying pluripotent cells to definitive endoderm. Mouse ESCs were encapsulated into heparin microgels with a single dose of Nodal and FGF-2, and expressed high levels of endoderm markers Sox17 and FoxA2 after 5 days. These results highlight the use of microencapsulation for tailoring the stem cell microenvironment to promote directed differentiation, and may provide a straightforward path to large scale bioprocessing in the future. STATEMENT OF SIGNIFICANCE Multicellular spheroids and microtissues are valuable for tissue engineering, but fabrication approaches typically sacrifice either precision or throughput. Microfluidic encapsulation in polymeric biomaterials is a promising technique for rapidly generating cell aggregates with excellent control of microenvironmental parameters. Here we describe the microfluidic fabrication of bioactive, heparin-based microgels, and demonstrate the adsorption of heparin-binding growth factors for enhancing directed differentiation of embryonic stem cells toward endoderm. This approach also facilitated a ∼90-fold decrease in consumption of exogenous growth factors compared to conventional differentiation protocols.
Collapse
|
27
|
Delgado-Coello B, Mas-Oliva J. Relevance of the plasma membrane calcium-ATPase in the homeostasis of calcium in the fetal liver. Organogenesis 2015; 10:333-9. [PMID: 25836032 PMCID: PMC4594366 DOI: 10.1080/15476278.2015.1011918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
During the early stages of development, the embryo depends on the placenta as provider of oxygen and calcium, among other essential compounds. Although fetal liver accomplishes a well-known haematopoietic function, its contribution to calcium homeostasis upon development is poorly understood. The homeostasis of cell calcium contributes to diverse signaling pathways across developmental stages of most tissues and the calcium-ATPase located at the plasma membrane (PMCA) helps pumping excess calcium into the extracellular space. To date, the understanding of the equilibrium shift between PMCA isoforms during liver development is still missing. This review focuses on the characterization of the hepatic PMCA along the early stages of development, followed by a description of modern approaches to study calcium homeostasis involving several types of pluripotent cells. The application of interdisciplinary techniques to improve our understanding of liver development and the role calcium homeostasis plays in the definition of pathogenesis is also discussed.
Collapse
Affiliation(s)
- Blanca Delgado-Coello
- a Departamento de Bioquímica y Biología Estructural ; Instituto de Fisiología Celular ; Universidad Nacional Autónoma de México ; México D.F. , México
| | | |
Collapse
|
28
|
Fagoonee S, Famulari ES, Silengo L, Tolosano E, Altruda F. Long Term Liver Engraftment of Functional Hepatocytes Obtained from Germline Cell-Derived Pluripotent Stem Cells. PLoS One 2015; 10:e0136762. [PMID: 26323094 PMCID: PMC4556379 DOI: 10.1371/journal.pone.0136762] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/08/2015] [Indexed: 12/25/2022] Open
Abstract
One of the major hurdles in liver gene and cell therapy is availability of ex vivo-expanded hepatocytes. Pluripotent stem cells are an attractive alternative. Here, we show that hepatocyte precursors can be isolated from male germline cell-derived pluripotent stem cells (GPSCs) using the hepatoblast marker, Liv2, and induced to differentiate into hepatocytes in vitro. These cells expressed hepatic-specific genes and were functional as demonstrated by their ability to secrete albumin and produce urea. When transplanted in the liver parenchyma of partially hepatectomised mice, Liv2-sorted cells showed regional and heterogeneous engraftment in the injected lobe. Moreover, approximately 50% of Y chromosome-positive, GPSC-derived cells were found in the female livers, in the region of engraftment, even one month after cell injection. This is the first study showing that Liv2-sorted GPSCs-derived hepatocytes can undergo long lasting engraftment in the mouse liver. Thus, GPSCs might offer promise for regenerative medicine.
Collapse
Affiliation(s)
- Sharmila Fagoonee
- Institute for Biostructures and Bioimages (CNR), Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- * E-mail: (SF); (FA)
| | - Elvira Smeralda Famulari
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Lorenzo Silengo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- * E-mail: (SF); (FA)
| |
Collapse
|
29
|
Lin CH, Hsiao YH, Chang HC, Yeh CF, He CK, Salm EM, Chen C, Chiu IM, Hsu CH. A microfluidic dual-well device for high-throughput single-cell capture and culture. LAB ON A CHIP 2015; 15:2928-38. [PMID: 26060987 DOI: 10.1039/c5lc00541h] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In vitro culture of single cells facilitates biological studies by deconvoluting complications from cell population heterogeneity. However, there is still a lack of simple yet high-throughput methods to perform single cell culture experiments. In this paper, we report the development and application of a microfluidic device with a dual-well (DW) design concept for high-yield single-cell loading (~77%) in large microwells (285 and 485 μm in diameter) which allowed for cell spreading, proliferation and differentiation. The increased single-cell loading yield is achieved by using sets of small microwells termed "capture-wells" and big microwells termed "culture-wells" according to their utilities for single-cell capture and culture, respectively. This novel device architecture allows the size of the "culture" microwells to be flexibly adjusted without affecting the single-cell loading efficiency making it useful for cell culture applications as demonstrated by our experiments of KT98 mouse neural stem cell differentiation, A549 and MDA-MB-435 cancer cell proliferation, and single-cell colony formation assay with A549 cells in this paper.
Collapse
Affiliation(s)
- Ching-Hui Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim JH, Jang YJ, An SY, Son J, Lee J, Lee G, Park JY, Park HJ, Hwang DY, Kim JH, Han J. Enhanced Metabolizing Activity of Human ES Cell-Derived Hepatocytes Using a 3D Culture System with Repeated Exposures to Xenobiotics. Toxicol Sci 2015; 147:190-206. [DOI: 10.1093/toxsci/kfv121] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
31
|
Siller R, Greenhough S, Naumovska E, Sullivan GJ. Small-molecule-driven hepatocyte differentiation of human pluripotent stem cells. Stem Cell Reports 2015; 4:939-52. [PMID: 25937370 PMCID: PMC4437467 DOI: 10.1016/j.stemcr.2015.04.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 12/17/2022] Open
Abstract
The differentiation of pluripotent stem cells to hepatocytes is well established, yet current methods suffer from several drawbacks. These include a lack of definition and reproducibility, which in part stems from continued reliance on recombinant growth factors. This has remained a stumbling block for the translation of the technology into industry and the clinic for reasons associated with cost and quality. We have devised a growth-factor-free protocol that relies on small molecules to differentiate human pluripotent stem cells toward a hepatic phenotype. The procedure can efficiently direct both human embryonic stem cells and induced pluripotent stem cells to hepatocyte-like cells. The final population of cells demonstrates marker expression at the transcriptional and protein levels, as well as key hepatic functions such as serum protein production, glycogen storage, and cytochrome P450 activity. Development of small-molecule-driven hepatocyte differentiation procedure for hPSCs Small-molecule-derived hepatocytes demonstrate key hepatic functions Significantly reduces the cost of hepatocyte differentiation Procedure is applicable to multiple human pluripotent stem cell lines
Collapse
Affiliation(s)
- Richard Siller
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Sebastian Greenhough
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Elena Naumovska
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Gareth J Sullivan
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway; Norwegian Center for Stem Cell Research, PO Box 1112 Blindern, 0317 Oslo, Norway; Institute of Immunology, Oslo University Hospital-Rikshospitalet, PO Box 4950 Nydalen, Oslo 0424, Norway.
| |
Collapse
|
32
|
Xu Y, Wang X. Fluid and cell behaviors along a 3D printed alginate/gelatin/fibrin channel. Biotechnol Bioeng 2015; 112:1683-95. [PMID: 25727058 DOI: 10.1002/bit.25579] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) cell manipulation is available with the integration of microfluidic technology and rapid prototyping techniques. High-Fidelity (Hi-Fi) constructs hold enormous therapeutic potential for organ manufacturing and regenerative medicine. In the present paper we introduced a quasi-three-dimensional (Q3D) model with parallel biocompatible alginate/gelatin/fibrin hurdles. The behaviors of fluids and cells along the microfluidic channels with various widths were studied. Cells inside the newly designed microfluidic channels attached and grew well. Morphological changes of adipose-derived stem cells (ADSCs) in both two-dimensional (2D) and 3D milieu were found on the printed constructs. Endothelialization occurred with the co-cultures of ADSCs and hepatocytes. This study provides insights into the interactions among fluids, cells and biomaterials, the behaviors of fluids and cells along the microfluidic channels, and the applications of Q3D techniques.
Collapse
Affiliation(s)
- Yufan Xu
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China
| | - Xiaohong Wang
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China. .,State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan, 430074, P.R. China.
| |
Collapse
|
33
|
Li S, Huang KJ, Wu JC, Hu MS, Sanyal M, Hu M, Longaker MT, Lorenz HP. Peripheral blood-derived mesenchymal stem cells: candidate cells responsible for healing critical-sized calvarial bone defects. Stem Cells Transl Med 2015; 4:359-68. [PMID: 25742693 DOI: 10.5966/sctm.2014-0150] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Postnatal tissue-specific stem/progenitor cells hold great promise to enhance repair of damaged tissues. Many of these cells are retrieved from bone marrow or adipose tissue via invasive procedures. Peripheral blood is an ideal alternative source for the stem/progenitor cells because of its ease of retrieval. We present a coculture system that routinely produces a group of cells from adult peripheral blood. Treatment with these cells enhanced healing of critical-size bone defects in the mouse calvarium, a proof of principle that peripheral blood-derived cells can be used to heal bone defects. From these cells, we isolated a subset of CD45(-) cells with a fibroblastic morphology. The CD45(-) cells were responsible for most of the differentiation-induced calcification activity and were most likely responsible for the enhanced healing process. These CD45(-) fibroblastic cells are plastic-adherent and exhibit a surface marker profile negative for CD34, CD19, CD11b, lineage, and c-kit and positive for stem cell antigen 1, CD73, CD44, CD90.1, CD29, CD105, CD106, and CD140α. Furthermore, these cells exhibited osteogenesis, chondrogenesis, and adipogenesis capabilities. The CD45(-) fibroblastic cells are the first peripheral blood-derived cells that fulfill the criteria of mesenchymal stem cells as defined by the International Society for Cellular Therapy. We have named these cells "blood-derived mesenchymal stem cells."
Collapse
Affiliation(s)
- Shaowei Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Ke-Jung Huang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Jen-Chieh Wu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael S Hu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Mrinmoy Sanyal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Min Hu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - H Peter Lorenz
- Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Pathology and Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
34
|
Choi H, Wadduwage DN, Tu TY, Matsudaira P, So PTC. Three-dimensional image cytometer based on widefield structured light microscopy and high-speed remote depth scanning. Cytometry A 2014; 87:49-60. [PMID: 25352187 DOI: 10.1002/cyto.a.22584] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/05/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022]
Abstract
A high throughput 3D image cytometer have been developed that improves imaging speed by an order of magnitude over current technologies. This imaging speed improvement was realized by combining several key components. First, a depth-resolved image can be rapidly generated using a structured light reconstruction algorithm that requires only two wide field images, one with uniform illumination and the other with structured illumination. Second, depth scanning is implemented using the high speed remote depth scanning. Finally, the large field of view, high NA objective lens and the high pixelation, high frame rate sCMOS camera enable high resolution, high sensitivity imaging of a large cell population. This system can image at 800 cell/sec in 3D at submicron resolution corresponding to imaging 1 million cells in 20 min. The statistical accuracy of this instrument is verified by quantitatively measuring rare cell populations with ratio ranging from 1:1 to 1:10(5) . © 2014 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Heejin Choi
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | | | | | | | | |
Collapse
|
35
|
Meneghello G, Storm MP, Chaudhuri JB, De Bank PA, Ellis MJ. An investigation into the stability of commercial versus MG63-derived hepatocyte growth factor under flow cultivation conditions. Biotechnol Lett 2014; 37:725-31. [PMID: 25331689 DOI: 10.1007/s10529-014-1701-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
The scale-up of tissue engineering cell culture must ensure that conditions are maintained while also being cost effective. Here we analyse the stability of hepatocyte growth factor (HGF) to investigate whether concentrations change under dynamic conditions, and compare commercial recombinant human HGF as an additive in 'standard medium', to HGF secreted by the osteosarcoma cell line MG63 as a 'preconditioned medium'. After 3 h under flow conditions, HGF in the standard medium degraded to 40% of its original concentration but HGF in the preconditioned medium remained at 100%. The concentration of secreted HGF was 10 times greater than the working concentration of commercially-available HGF. Thus HGF within this medium has increased stability; MG63-derived HGF should therefore be investigated as a cost-effective alternative to current lyophilised powders for use in in vitro models. Furthermore, we recommend that those intending to use HGF (or other growth factors) should consider similar stability testing before embarking on experiments with media flow.
Collapse
Affiliation(s)
- Giulia Meneghello
- Department of Pharmacy and Pharmacology, Centre for Regenerative Medicine, University of Bath, Bath, BA2 7AY, UK
| | | | | | | | | |
Collapse
|
36
|
Kadota Y, Yagi H, Inomata K, Matsubara K, Hibi T, Abe Y, Kitago M, Shinoda M, Obara H, Itano O, Kitagawa Y. Mesenchymal stem cells support hepatocyte function in engineered liver grafts. Organogenesis 2014; 10:268-77. [PMID: 24488046 DOI: 10.4161/org.27879] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Recent studies suggest that organ decellularization is a promising approach to facilitate the clinical application of regenerative therapy by providing a platform for organ engineering. This unique strategy uses native matrices to act as a reservoir for the functional cells which may show therapeutic potential when implanted into the body. Appropriate cell sources for artificial livers have been debated for some time. The desired cell type in artificial livers is primary hepatocytes, but in addition, other supportive cells may facilitate this stem cell technology. In this context, the use of mesenchymal stem cells (MSC) is an option meeting the criteria for therapeutic organ engineering. Ideally, supportive cells are required to (1) reduce the hepatic cell mass needed in an engineered liver by enhancing hepatocyte function, (2) modulate hepatic regeneration in a paracrine fashion or by direct contact, and (3) enhance the preservability of parenchymal cells during storage. Here, we describe enhanced hepatic function achieved using a strategy of sequential infusion of cells and illustrate the advantages of co-cultivating bone marrow-derived MSCs with primary hepatocytes in the engineered whole-liver scaffold. These co-recellularized liver scaffolds colonized by MSCs and hepatocytes were transplanted into live animals. After blood flow was established, we show that expression of adhesion molecules and proangiogenic factors was upregulated in the graft.
Collapse
Affiliation(s)
- Yoshie Kadota
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Kenta Inomata
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Kentaro Matsubara
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Taizo Hibi
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Yuta Abe
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Masahiro Shinoda
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Hideaki Obara
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Osamu Itano
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery; Keio University; School of Medicine; Tokyo, Japan
| |
Collapse
|
37
|
Handa K, Matsubara K, Fukumitsu K, Guzman-Lepe J, Watson A, Soto-Gutierrez A. Assembly of human organs from stem cells to study liver disease. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:348-57. [PMID: 24333262 DOI: 10.1016/j.ajpath.2013.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/04/2013] [Accepted: 11/18/2013] [Indexed: 01/01/2023]
Abstract
Recently, significant developments in the field of liver tissue engineering have raised new possibilities for the study of complex physiological and pathophysiological processes in vitro, as well as the potential to assemble entire organs for transplantation. Human-induced pluripotent stem cells have been differentiated into relatively functional populations of hepatic cells, and novel techniques to generate whole organ acellular three-dimensional scaffolds have been developed. In this review, we highlight the most recent advances in organ assembly regarding the development of liver tissue in vitro. We emphasize applications that involve multiple types of cells with a biomimetic spatial organization for which three-dimensional configurations could be used for drug development or to explain mechanisms of disease. We also discuss applications of liver organotypic surrogates and the challenges of translating the highly promising new field of tissue engineering into a proven platform for predicting drug metabolism and toxicity.
Collapse
Affiliation(s)
- Kan Handa
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kentaro Matsubara
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ken Fukumitsu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jorge Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alicia Watson
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
38
|
Dianat N, Steichen C, Vallier L, Weber A, Dubart-Kupperschmitt A. Human pluripotent stem cells for modelling human liver diseases and cell therapy. Curr Gene Ther 2013; 13:120-32. [PMID: 23444872 PMCID: PMC3882648 DOI: 10.2174/1566523211313020006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 12/24/2022]
Abstract
The liver is affected by many types of diseases, including metabolic disorders and acute liver failure. Orthotopic liver transplantation (OLT) is currently the only effective treatment for life-threatening liver diseases but transplantation of allogeneic hepatocytes has now become an alternative as it is less invasive than OLT and can be performed repeatedly. However, this approach is hampered by the shortage of organ donors, and the problems related to the isolation of high quality adult hepatocytes, their cryopreservation and their absence of proliferation in culture. Liver is also a key organ to assess the pharmacokinetics and toxicology of xenobiotics and for drug discovery, but appropriate cell culture systems are lacking. All these problems have highlighted the need to explore other sources of cells such as stem cells that could be isolated, expanded to yield sufficiently large populations and then induced to differentiate into functional hepatocytes. The presence of a niche of “facultative” progenitor and stem cells in the normal liver has recently been confirmed but they display no telomerase activity. The recent discovery that human induced pluripotent stem cells can be generated from somatic cells has renewed hopes for regenerative medicine and in vitro disease modelling, as these cells are easily accessible. We review here the present progresses, limits and challenges for the generation of functional hepatocytes from human pluripotent stem cells in view of their potential use in regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Noushin Dianat
- INSERM UMR-S972, Paul Brousse Hospital, Villejuif, F-94807, France
| | | | | | | | | |
Collapse
|
39
|
Qian NS, Liu WH, Lv WP, Xiang X, Su M, Raut V, Chen YL, Dong JH. Upregulated microRNA-92b regulates the differentiation and proliferation of EpCAM-positive fetal liver cells by targeting C/EBPß. PLoS One 2013; 8:e68004. [PMID: 23936298 PMCID: PMC3732262 DOI: 10.1371/journal.pone.0068004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 05/29/2013] [Indexed: 12/15/2022] Open
Abstract
microRNAs (miRNAs) are short noncoding RNAs that negatively regulate gene expression. Although recent evidences have been indicated that their aberrant expression may play an important role in cancer stem cells, the mechanism of their deregulation in neoplastic transformation of liver cancer stem cells (LCSCs) has not been explored. In our study, the HCC model was established in F344 rats by DEN induction. The EpCAM+ cells were sorted out from unfractionated fetal liver cells and liver cancer cells using the FACS analysis and miRNA expression profiles of two groups were screened through microarray platform. Gain-of-function studies were performed in vitro and in vivo to determine the role of miR-92b on proliferation and differentiation of the hepatic progenitors. In addition, luciferase reporter system and gene function analysis were used to predict miR-92b target. we found that miR-92b was highly downregulated in EpCAM+ fetal liver cells in expression profiling studies. RT-PCR analysis demonstrated reverse correlation between miR-92b expression and differentiation degree in human HCC samples. Overexpression of miR-92b in EpCAM+ fetal liver cells significantly increased proliferation and inhibited differentiation as well as in vitro and in vivo studies. Moreover, we verified that C/EBPß is a direct target of miR-92b and contributes to its effects on proliferation and differentiation. We conclude that aberrant expression of miR-92b can result in proliferation increase and differentiation arrest of hepatic progenitors by targeting C/EBPß.
Collapse
Affiliation(s)
- Nian-Song Qian
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
- Department of Hepatobiliary Surgery, Hainan Branch of PLA General Hospital, Sanya, China
- * E-mail: (YLC); (NSQ)
| | - Wei-Hui Liu
- Department of Hepatobiliary Surgery, General Hospital of Chengdu Military Region, Chengdu, China
| | - Wen-Ping Lv
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
- Department of Hepatobiliary Surgery, Hainan Branch of PLA General Hospital, Sanya, China
| | - Xin Xiang
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
- Department of Hepatobiliary Surgery, Hainan Branch of PLA General Hospital, Sanya, China
| | - Ming Su
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
- Department of Hepatobiliary Surgery, Hainan Branch of PLA General Hospital, Sanya, China
| | - Vikram Raut
- Department of Hepatobiliary Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yong-Liang Chen
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
- Department of Hepatobiliary Surgery, Hainan Branch of PLA General Hospital, Sanya, China
- * E-mail: (YLC); (NSQ)
| | - Jia-Hong Dong
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
| |
Collapse
|
40
|
Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, Braeuning A, Budinsky RA, Burkhardt B, Cameron NR, Camussi G, Cho CS, Choi YJ, Craig Rowlands J, Dahmen U, Damm G, Dirsch O, Donato MT, Dong J, Dooley S, Drasdo D, Eakins R, Ferreira KS, Fonsato V, Fraczek J, Gebhardt R, Gibson A, Glanemann M, Goldring CEP, Gómez-Lechón MJ, Groothuis GMM, Gustavsson L, Guyot C, Hallifax D, Hammad S, Hayward A, Häussinger D, Hellerbrand C, Hewitt P, Hoehme S, Holzhütter HG, Houston JB, Hrach J, Ito K, Jaeschke H, Keitel V, Kelm JM, Kevin Park B, Kordes C, Kullak-Ublick GA, LeCluyse EL, Lu P, Luebke-Wheeler J, Lutz A, Maltman DJ, Matz-Soja M, McMullen P, Merfort I, Messner S, Meyer C, Mwinyi J, Naisbitt DJ, Nussler AK, Olinga P, Pampaloni F, Pi J, Pluta L, Przyborski SA, Ramachandran A, Rogiers V, Rowe C, Schelcher C, Schmich K, Schwarz M, Singh B, Stelzer EHK, Stieger B, Stöber R, Sugiyama Y, Tetta C, Thasler WE, Vanhaecke T, Vinken M, Weiss TS, Widera A, Woods CG, Xu JJ, Yarborough KM, Hengstler JG. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013; 87:1315-530. [PMID: 23974980 PMCID: PMC3753504 DOI: 10.1007/s00204-013-1078-5] [Citation(s) in RCA: 1062] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
This review encompasses the most important advances in liver functions and hepatotoxicity and analyzes which mechanisms can be studied in vitro. In a complex architecture of nested, zonated lobules, the liver consists of approximately 80 % hepatocytes and 20 % non-parenchymal cells, the latter being involved in a secondary phase that may dramatically aggravate the initial damage. Hepatotoxicity, as well as hepatic metabolism, is controlled by a set of nuclear receptors (including PXR, CAR, HNF-4α, FXR, LXR, SHP, VDR and PPAR) and signaling pathways. When isolating liver cells, some pathways are activated, e.g., the RAS/MEK/ERK pathway, whereas others are silenced (e.g. HNF-4α), resulting in up- and downregulation of hundreds of genes. An understanding of these changes is crucial for a correct interpretation of in vitro data. The possibilities and limitations of the most useful liver in vitro systems are summarized, including three-dimensional culture techniques, co-cultures with non-parenchymal cells, hepatospheres, precision cut liver slices and the isolated perfused liver. Also discussed is how closely hepatoma, stem cell and iPS cell-derived hepatocyte-like-cells resemble real hepatocytes. Finally, a summary is given of the state of the art of liver in vitro and mathematical modeling systems that are currently used in the pharmaceutical industry with an emphasis on drug metabolism, prediction of clearance, drug interaction, transporter studies and hepatotoxicity. One key message is that despite our enthusiasm for in vitro systems, we must never lose sight of the in vivo situation. Although hepatocytes have been isolated for decades, the hunt for relevant alternative systems has only just begun.
Collapse
Affiliation(s)
- Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | | | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Nariman Ansari
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Sudin Bhattacharya
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jennifer Bolleyn
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Jan Böttger
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Albert Braeuning
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Robert A. Budinsky
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Neil R. Cameron
- Department of Chemistry, Durham University, Durham, DH1 3LE UK
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - J. Craig Rowlands
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General Visceral, and Vascular Surgery, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Olaf Dirsch
- Institute of Pathology, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - María Teresa Donato
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jian Dong
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dirk Drasdo
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
- INRIA (French National Institute for Research in Computer Science and Control), Domaine de Voluceau-Rocquencourt, B.P. 105, 78153 Le Chesnay Cedex, France
- UPMC University of Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, 4, pl. Jussieu, 75252 Paris cedex 05, France
| | - Rowena Eakins
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Karine Sá Ferreira
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- GRK 1104 From Cells to Organs, Molecular Mechanisms of Organogenesis, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Valentina Fonsato
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Joanna Fraczek
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Andrew Gibson
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Matthias Glanemann
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| | - Geny M. M. Groothuis
- Department of Pharmacy, Pharmacokinetics Toxicology and Targeting, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lena Gustavsson
- Department of Laboratory Medicine (Malmö), Center for Molecular Pathology, Lund University, Jan Waldenströms gata 59, 205 02 Malmö, Sweden
| | - Christelle Guyot
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Seddik Hammad
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Adam Hayward
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Claus Hellerbrand
- Department of Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
| | - Hermann-Georg Holzhütter
- Institut für Biochemie Abteilung Mathematische Systembiochemie, Universitätsmedizin Berlin (Charité), Charitéplatz 1, 10117 Berlin, Germany
| | - J. Brian Houston
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | | | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585 Japan
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claus Kordes
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Peng Lu
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | - Anna Lutz
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Daniel J. Maltman
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
| | - Madlen Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas K. Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jingbo Pi
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Stefan A. Przyborski
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Vera Rogiers
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cliff Rowe
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Celine Schelcher
- Department of Surgery, Liver Regeneration, Core Facility, Human in Vitro Models of the Liver, Ludwig Maximilians University of Munich, Munich, Germany
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Ernst H. K. Stelzer
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Regina Stöber
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama Biopharmaceutical R&D Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Wolfgang E. Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, Munich, Germany
| | - Tamara Vanhaecke
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas S. Weiss
- Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Agata Widera
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Courtney G. Woods
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| |
Collapse
|
41
|
Chan KM, Fu YH, Wu TJ, Hsu PY, Lee WC. Hepatic stellate cells promote the differentiation of embryonic stem cell-derived definitive endodermal cells into hepatic progenitor cells. Hepatol Res 2013; 43:648-57. [PMID: 23072626 DOI: 10.1111/j.1872-034x.2012.01110.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 09/14/2012] [Accepted: 09/17/2012] [Indexed: 02/08/2023]
Abstract
AIM Hepatic non-parenchymal cells are well known to be capable of providing an important microenvironment and growth factors for hepatic regeneration, but their capacity for directing embryonic stem cells (ESC) toward hepatocytes remains to be assessed. Thus, this study aims to investigate the role of hepatic stellate cells (HSC), the major type of hepatic non-parenchymal cells, in the differentiation of ESC as well as exploring the potentiality of ESC in regeneration medicine for cell-based therapy. METHODS A two-step differentiation procedure that utilized the capability of HSC to regulate proliferation and differentiation of hepatocytes was used to develop an approach for directing the differentiation of ESC towards hepatic progenitor cells. Mouse ESC were cultivated in a serum-free medium containing Activin A and fibroblast growth factor to generate definitive endodermal cells characterized by the CXCR4 cell-surface marker. After 6-8 days in culture, approximately 60% of the differentiated cells expressed CXCR4, and more than 90% of the CXCR4 positive cells could be recovered by cell sorting. The purified CXCR4 positive cells were co-cultured with mouse HSC as feeder cells in basal medium without additional hepatocyte growth factors. Differentiation was complete after 10-12 days of co-culture, and hepatic progenitor cell markers such as α-fetoprotein (afp) and albumin (alb) were detected in the terminally differentiated ESC. CONCLUSION These results show that HSC provide an appropriate microenvironment and pivotal growth factors for generation of hepatic progenitor cells from ESC-derived definitive endodermal cells, and suggest that this approach possibly allows for hepatic differentiation of ESC imitating the process of hepatic regeneration.
Collapse
Affiliation(s)
- Kun-Ming Chan
- Division of Liver and Transplantation Surgery, Department of General Surgery, Chang-Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | | | | | | | | |
Collapse
|
42
|
Abstract
Liver is a prime organ responsible for synthesis, metabolism, and detoxification. The organ is endodermal in origin and its development is regulated by temporal, complex, and finely balanced cellular and molecular interactions that dictate its origin, growth, and maturation. We discuss the relevance of endoderm patterning, which truly is the first step toward mapping of domains that will give rise to specific organs. Once foregut patterning is completed, certain cells within the foregut endoderm gain competence in the form of expression of certain transcription factors that allow them to respond to certain inductive signals. Hepatic specification is then a result of such inductive signals, which often emanate from the surrounding mesenchyme. During hepatic specification bipotential hepatic stem cells or hepatoblasts become apparent and undergo expansion, which results in a visible liver primordium during the stage of hepatic morphogenesis. Hepatoblasts next differentiate into either hepatocytes or cholangiocytes. The expansion and differentiation is regulated by cellular and molecular interactions between hepatoblasts and mesenchymal cells including sinusoidal endothelial cells, stellate cells, and also innate hematopoietic elements. Further maturation of hepatocytes and cholangiocytes continues during late hepatic development as a function of various growth factors. At this time, liver gains architectural novelty in the form of zonality and at cellular level acquires polarity. A comprehensive elucidation of such finely tuned developmental cues have been the basis of transdifferentiation of various types of stem cells to hepatocyte-like cells for purposes of understanding health and disease and for therapeutic applications.
Collapse
Affiliation(s)
- Donghun Shin
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | |
Collapse
|
43
|
Bajaj P, Marchwiany D, Duarte C, Bashir R. Patterned three-dimensional encapsulation of embryonic stem cells using dielectrophoresis and stereolithography. Adv Healthc Mater 2013; 2:450-8. [PMID: 23463644 DOI: 10.1002/adhm.201200318] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 11/03/2012] [Indexed: 01/07/2023]
Abstract
Controlling the assembly of cells in three dimensions is very important for engineering functional tissues, drug screening, probing cell-cell/cell-matrix interactions, and studying the emergent behavior of cellular systems. Although the current methods of cell encapsulation in hydrogels can distribute them in three dimensions, these methods typically lack spatial control of multi-cellular organization and do not allow for the possibility of cell-cell contacts as seen for the native tissue. Here, we report the integration of dielectrophoresis (DEP) with stereolithography (SL) apparatus for the spatial patterning of cells on custom made gold micro-electrodes. Afterwards, they are encapsulated in poly (ethylene glycol) diacrylate (PEGDA) hydrogels of different stiffnesses. This technique can mimic the in vivo microscale tissue architecture, where the cells have a high degree of three dimensional (3D) spatial control. As a proof of concept, we show the patterning and encapsulation of mouse embryonic stem cells (mESCs) and C2C12 skeletal muscle myoblasts. mESCs show high viability in both the DEP (91.79% ± 1.4%) and the no DEP (94.27% ± 0.5%) hydrogel samples. Furthermore, we also show the patterning of mouse embryoid bodies (mEBs) and C2C12 spheroids in the hydrogels, and verify their viability. This robust and flexible in vitro platform can enable various applications in stem cell differentiation and tissue engineering by mimicking elements of the native 3D in vivo cellular micro-environment.
Collapse
Affiliation(s)
- Piyush Bajaj
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
44
|
Park SJ, Bae D, Moon SH, Chung HM. Modification of a purification and expansion method for human embryonic stem cell-derived cardiomyocytes. Cardiology 2013; 124:139-50. [PMID: 23428747 DOI: 10.1159/000346390] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 11/30/2012] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study aimed to develop a simple and efficient purification method for human embryonic stem cell (hESC)-derived cardiomyocytes (CMs) using a low-glucose culture system. In addition, we investigated whether intercellular adhesion between single hESC-CMs plays a critical role in enhancing proliferation of purified hESC-CMs. METHOD hESCs were cultured in suspension to form human embryoid bodies (hEBs) from which ∼15% contracting clusters were derived after 15-20 days in culture. To purify CMs from contracting hEBs, we first manually isolated contracting clumps that were re-cultured on gelatin-coated plates with media containing a low concentration of glucose. The purified hESC-CMs were cultured at different densities to examine whether cell-cell contact enhances proliferation of hESC-CMs. RESULTS Purified CMs demonstrated spontaneous contraction and strongly expressed the CM-specific markers cardiac troponin T and slow myosin heavy chain. We investigated the purification efficiency by examining the expression levels of cardiac-related genes and the expression of MitoTracker Red dye. In addition, purified hESC-CMs in low-glucose culture demonstrated a 1.5-fold increase in their proliferative capacity compared to those cultured as single hESC-CMs. CONCLUSION A low level of glucose is efficient in purifying hESC-CMs and intercellular adhesion between individual hESC-CMs plays a critical role in enhancing hESC-CM proliferation.
Collapse
Affiliation(s)
- Soon-Jung Park
- Stem Cell Research Laboratory, CHA Stem Cell Institute, CHA University, Seol 135-081, Korea
| | | | | | | |
Collapse
|
45
|
Iwamuro M, Shiraha H, Nakaji S, Furutani M, Kobayashi N, Takaki A, Yamamoto K. A preliminary study for constructing a bioartificial liver device with induced pluripotent stem cell-derived hepatocytes. Biomed Eng Online 2012; 11:93. [PMID: 23217363 PMCID: PMC3549893 DOI: 10.1186/1475-925x-11-93] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 12/04/2012] [Indexed: 12/21/2022] Open
Abstract
Background Bioartificial liver systems, designed to support patients with liver failure, are composed of bioreactors and functional hepatocytes. Immunological rejection of the embedded hepatocytes by the host immune system is a serious concern that crucially degrades the performance of the device. Induced pluripotent stem (iPS) cells are considered a desirable source for bioartificial liver systems, because patient-derived iPS cells are free from immunological rejection. The purpose of this paper was to test the feasibility of a bioartificial liver system with iPS cell-derived hepatocyte-like cells. Methods Mouse iPS cells were differentiated into hepatocyte-like cells by a multi-step differentiation protocol via embryoid bodies and definitive endoderm. Differentiation of iPS cells was evaluated by morphology, PCR assay, and functional assays. iPS cell-derived hepatocyte-like cells were cultured in a bioreactor module with a pore size of 0.2 μm for 7 days. The amount of albumin secreted into the circulating medium was analyzed by ELISA. Additionally, after a 7-day culture in a bioreactor module, cells were observed by a scanning electron microscope. Results At the final stage of the differentiation program, iPS cells changed their morphology to a polygonal shape with two nucleoli and enriched cytoplasmic granules. Transmission electron microscope analysis revealed their polygonal shape, glycogen deposition in the cytoplasm, microvilli on their surfaces, and a duct-like arrangement. PCR analysis showed increased expression of albumin mRNA over the course of the differentiation program. Albumin and urea production was also observed. iPS-Heps culture in bioreactor modules showed the accumulation of albumin in the medium for up to 7 days. Scanning electron microscopy revealed the attachment of cell clusters to the hollow fibers of the module. These results indicated that iPS cells were differentiated into hepatocyte-like cells after culture for 7 days in a bioreactor module with a pore size of 0.2 μm. Conclusion We consider the combination of a bioreactor module with a 0.2-μm pore membrane and embedded hepatocytes differentiated from iPS cells to be a promising option for bioartificial liver systems. This paper provides the basic concept and preliminary data for an iPS cell-oriented bioartificial liver system. PACS code: 87. Biological and medical physics, 87.85.-d Biomedical engineering, 87.85.Lf Tissue engineering, 87.85.Tu Modeling biomedical systems.
Collapse
Affiliation(s)
- Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Soto-Gutierrez A, Wertheim JA, Ott HC, Gilbert TW. Perspectives on whole-organ assembly: moving toward transplantation on demand. J Clin Invest 2012; 122:3817-23. [PMID: 23114604 DOI: 10.1172/jci61974] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There is an ever-growing demand for transplantable organs to replace acute and chronically damaged tissues. This demand cannot be met by the currently available donor organs. Efforts to provide an alternative source have led to the development of organ engineering, a discipline that combines cell biology, tissue engineering, and cell/organ transplantation. Over the last several years, engineered organs have been implanted into rodent recipients and have shown modest function. In this article, we summarize the most recent advances in this field and provide a perspective on the challenges of translating this promising new technology into a proven regenerative therapy.
Collapse
Affiliation(s)
- Alejandro Soto-Gutierrez
- Department of Pathology, Transplantation Section of Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | | | |
Collapse
|
47
|
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]
|
48
|
Liu WH, Liu ZC, You N, Zhang N, Wang T, Gong ZB, Liu HB, Dou KF. Several important in vitro improvements in the amplification, differentiation and tracing of fetal liver stem/progenitor cells. PLoS One 2012; 7:e47346. [PMID: 23056632 PMCID: PMC3467257 DOI: 10.1371/journal.pone.0047346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/11/2012] [Indexed: 01/12/2023] Open
Abstract
Objective We previously isolated fetal liver stem/progenitor cells (FLSPCs), but there is an urgent need to properly amplify FLSPCs, effectively induce FLSPCs differentiation, and steadily trace FLSPCs for in vivo therapeutic investigation. Methods FLSPCs were maintained in vitro as adherent culture or soft agar culture for large-scale amplification. To direct the differentiation of FLSPCs into hepatocytes, FLSPCs were randomly divided into four groups: control, 1% DMSO-treated, 20 ng/ml HGF-treated and 1% DMSO+20 ng/ml HGF-treated. To trace FLSPCs, the GFP gene was introduced into FLSPCs by liposome-mediated transfection. Results For amplifying FLSPCs, the soft agar culture were more suitable than the adherent culture, because the soft agar culture obtained more homogeneous cells. These cells were with high nuclear:cytoplasmic ratio, few cell organelles, high expression of CD90.1 and CD49f, and strong alkaline phosphatase staining. For inducing FLSPCs differentiation, treatment with HGF+DMSO was most effective (P<0.05), which was strongly supported by the typical morphological change and the significant decrease of OV-6 positive cells (P<0.01). In addition, the time of indocyanine green elimination, the percentage of glycogen synthetic cells, and the expressions of ALB, G-6-P, CK-8, CK-18 and CYP450-3A1 in HGF+DMSO-treated group were higher than in any other group. For tracing FLSPCs, after the selection of stable FLSPC transfectants, GFP expression continued over successive generations. Conclusions FLSPCs can properly self-renew in soft agar culture and effectively differentiate into hepatocyte-like cells by HGF+DMSO induction, and they can be reliably traced by GFP expression.
Collapse
Affiliation(s)
- Wei-hui Liu
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, People's Republic of China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Zheng-cai Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan You
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Ning Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Tao Wang
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, People's Republic of China
| | - Zhen-bin Gong
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Hong-bao Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
- * E-mail: (K-fD); (H-bL)
| | - Ke-feng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
- * E-mail: (K-fD); (H-bL)
| |
Collapse
|
49
|
Nucleic acid and non-nucleic acid-based reprogramming of adult limbal progenitors to pluripotency. PLoS One 2012; 7:e46734. [PMID: 23056428 PMCID: PMC3466310 DOI: 10.1371/journal.pone.0046734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/31/2012] [Indexed: 01/21/2023] Open
Abstract
Reprogramming somatic cells to a pluripotent state by nucleic acid based (NAB) approaches, involving the ectopic expression of transcription factors, has emerged as a standard method. We recently demonstrated that limbal progenitors that regenerate cornea are reprogrammable to pluripotency by a non-NAB approach through simple manipulation of microenvironment thus extending the possible therapeutic use of these readily accessible cells beyond the proven treatment of corneal diseases and injury. Therefore, to determine the validity and robustness of non-cell autonomous reprogramming of limbal progenitors for a wider clinical use, here, we have compared their reprogramming by non-NAB and NAB approaches. We observed that both approaches led to (1) the emergence of colonies displaying pluripotency markers, accompanied by a temporal reciprocal changes in limbal-specific and pluripotency gene expression, and (2) epigenetic alterations of Oct4 and Nanog, associated with the de-novo activation of their expression. While the efficiency of reprogramming and passaging of re-programmed cells were significantly better with the NAB approach, the non-NAB approach, in contrast, led to a regulated reprogramming of gene expression, and a significant decrease in the expression of Hormad1, a gene associated with immunogenic responses. The reprogramming efficiency by non-NAB approach was influenced by exosomes present in conditioned medium. Cells reprogrammed by both approaches were capable of differentiating along the three germ lineages and generating chimeras. The analysis suggests that both approaches are effective in reprogramming limbal progenitors but the non-NAB approach may be more suitable for potential clinical applications by averting the risk of insertional mutagenesis and immune responses associated with the NAB approach.
Collapse
|
50
|
Wash R, Calabressi S, Franz S, Griffiths SJ, Goulding D, Tan EP, Wise H, Digard P, Haas J, Efstathiou S, Kellam P. Permissive and restricted virus infection of murine embryonic stem cells. J Gen Virol 2012; 93:2118-2130. [PMID: 22815272 PMCID: PMC3541792 DOI: 10.1099/vir.0.043406-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/13/2012] [Indexed: 12/13/2022] Open
Abstract
Recent RNA interference (RNAi) studies have identified many host proteins that modulate virus infection, but small interfering RNA 'off-target' effects and the use of transformed cell lines limit their conclusiveness. As murine embryonic stem (mES) cells can be genetically modified and resources exist where many and eventually all known mouse genes are insertionally inactivated, it was reasoned that mES cells would provide a useful alternative to RNAi screens. Beyond allowing investigation of host-pathogen interactions in vitro, mES cells have the potential to differentiate into other primary cell types, as well as being used to generate knockout mice for in vivo studies. However, mES cells are poorly characterized for virus infection. To investigate whether ES cells can be used to explore host-virus interactions, this study characterized the responses of mES cells following infection by herpes simplex virus type 1 (HSV-1) and influenza A virus. HSV-1 replicated lytically in mES cells, although mES cells were less permissive than most other cell types tested. Influenza virus was able to enter mES cells and express some viral proteins, but the replication cycle was incomplete and no infectious virus was produced. Knockdown of the host protein AHCYL1 in mES cells reduced HSV-1 replication, showing the potential for using mES cells to study host-virus interactions. Transcriptional profiling, however, indicated the lack of an efficient innate immune response in these cells. mES cells may thus be useful to identify host proteins that play a role in virus replication, but they are not suitable to determine factors that are involved in innate host defence.
Collapse
Affiliation(s)
- Rachael Wash
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Sabrina Calabressi
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Stephanie Franz
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Samantha J. Griffiths
- Division of Pathway Medicine, The University of Edinburgh, Old College, South Bridge, Edinburgh, EH8 9YL, UK
| | - David Goulding
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - E-Pien Tan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Helen Wise
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Paul Digard
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Jürgen Haas
- Division of Pathway Medicine, The University of Edinburgh, Old College, South Bridge, Edinburgh, EH8 9YL, UK
| | - Stacey Efstathiou
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Paul Kellam
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- UCL/MRC Centre for Medical Molecular Virology, Department of Infection, University College London, London WC1E 6BT, UK
| |
Collapse
|