1
|
Liu J, Hanson A, Yin W, Wu Q, Wauthier E, Diao J, Dinh T, Macdonald J, Li R, Terajima M, Yamauchi M, Chen Z, Sethupathy P, Dong J, Reid LM, Wang Y. Decellularized liver scaffolds for constructing drug-metabolically functional ex vivo human liver models. Bioact Mater 2025; 43:162-180. [PMID: 39386220 PMCID: PMC11462156 DOI: 10.1016/j.bioactmat.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
The creation of ex vivo human liver models has long been a critical objective in academic, clinical, and pharmaceutical research, particularly for drug development, where accurate evaluation of hepatic metabolic dynamics is crucial. We have developed a bioengineered, perfused, organ-level human liver model that accurately replicates key liver functions, including metabolic activities, and protein synthesis, thus addressing some of the limitations associated with traditional liver monolayers, organoids, and matrix-embedded liver cells. Our approach utilizes liver-specific biomatrix scaffolds, prepared using an innovative protocol and fortified with matrix components that facilitate cellular interactions. These scaffolds, when seeded with human fetal liver cells or co-seeded with liver parenchymal and endothelial cell lines, enable the formation of three-dimensional (3D) human livers with enhanced cellular organization. The "recellularized tissue-engineered livers" (RCLs) have undergone various analyses, demonstrating the capability for establishing liver microenvironments ex vivo. Within 7-14 days, the RCLs exhibit evidence of liver differentiation and metabolic capabilities, underscoring the potential for use in drug metabolism and toxicity studies. Although our study represents a significant step forward, we acknowledge the need for direct comparisons with existing models and further research to fully elucidate the spectrum of regenerative responses. The high drug-metabolizing enzyme activity of RCLs, as demonstrated in our study, provides a promising avenue for investigating drug-induced liver injury mechanisms, contributing to a more detailed understanding of early drug discovery processes.
Collapse
Affiliation(s)
- Juan Liu
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Ariel Hanson
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Wenzhen Yin
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Qiao Wu
- Infection Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Eliane Wauthier
- Departments of Cell Biology and Physiology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jinmei Diao
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Timothy Dinh
- Departments of Genetics, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jeff Macdonald
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Ruihong Li
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Masahiko Terajima
- Oral and Craniofacial Health Sciences, UNC School of Dentistry, Chapel Hill, NC, 27599, USA
| | - Mitsuo Yamauchi
- Oral and Craniofacial Health Sciences, UNC School of Dentistry, Chapel Hill, NC, 27599, USA
| | - Ziye Chen
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Praveen Sethupathy
- Departments of Genetics, UNC School of Medicine, Chapel Hill, NC, 27599, USA
- Division of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Jiahong Dong
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Lola M. Reid
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
- Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Yunfang Wang
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
2
|
Zhang W, Cui Y, Du Y, Yang Y, Fang T, Lu F, Kong W, Xiao C, Shi J, Reid LM, He Z. Liver cell therapies: cellular sources and grafting strategies. Front Med 2023; 17:432-457. [PMID: 37402953 DOI: 10.1007/s11684-023-1002-1] [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: 01/17/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023]
Abstract
The liver has a complex cellular composition and a remarkable regenerative capacity. The primary cell types in the liver are two parenchymal cell populations, hepatocytes and cholangiocytes, that perform most of the functions of the liver and that are helped through interactions with non-parenchymal cell types comprising stellate cells, endothelia and various hemopoietic cell populations. The regulation of the cells in the liver is mediated by an insoluble complex of proteins and carbohydrates, the extracellular matrix, working synergistically with soluble paracrine and systemic signals. In recent years, with the rapid development of genetic sequencing technologies, research on the liver's cellular composition and its regulatory mechanisms during various conditions has been extensively explored. Meanwhile breakthroughs in strategies for cell transplantation are enabling a future in which there can be a rescue of patients with end-stage liver diseases, offering potential solutions to the chronic shortage of livers and alternatives to liver transplantation. This review will focus on the cellular mechanisms of liver homeostasis and how to select ideal sources of cells to be transplanted to achieve liver regeneration and repair. Recent advances are summarized for promoting the treatment of end-stage liver diseases by forms of cell transplantation that now include grafting strategies.
Collapse
Affiliation(s)
- Wencheng Zhang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yangyang Cui
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
- Postgraduate Training Base of Shanghai East Hospital, Jinzhou Medical University, Jinzhou, 121001, China
| | - Yuan Du
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yong Yang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Ting Fang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Fengfeng Lu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Weixia Kong
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Canjun Xiao
- Department of General Surgery, Ji'an Hospital, Shanghai East Hospital, School of Medicine, Tongji University, Ji'an, 343006, China
| | - Jun Shi
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of General Surgery, Ji'an Hospital, Shanghai East Hospital, School of Medicine, Tongji University, Ji'an, 343006, China
| | - Lola M Reid
- Department of Cell Biology and Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA.
| | - Zhiying He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, China.
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
3
|
Schönberger K, Mitterer M, Glaser K, Stecher M, Hobitz S, Schain-Zota D, Schuldes K, Lämmermann T, Rambold AS, Cabezas-Wallscheid N, Buescher JM. LC-MS-Based Targeted Metabolomics for FACS-Purified Rare Cells. Anal Chem 2023; 95:4325-4334. [PMID: 36812587 PMCID: PMC9996616 DOI: 10.1021/acs.analchem.2c04396] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Metabolism plays a fundamental role in regulating cellular functions and fate decisions. Liquid chromatography-mass spectrometry (LC-MS)-based targeted metabolomic approaches provide high-resolution insights into the metabolic state of a cell. However, the typical sample size is in the order of 105-107 cells and thus not compatible with rare cell populations, especially in the case of a prior flow cytometry-based purification step. Here, we present a comprehensively optimized protocol for targeted metabolomics on rare cell types, such as hematopoietic stem cells and mast cells. Only 5000 cells per sample are required to detect up to 80 metabolites above background. The use of regular-flow liquid chromatography allows for robust data acquisition, and the omission of drying or chemical derivatization avoids potential sources of error. Cell-type-specific differences are preserved while the addition of internal standards, generation of relevant background control samples, and targeted metabolite with quantifiers and qualifiers ensure high data quality. This protocol could help numerous studies to gain thorough insights into cellular metabolic profiles and simultaneously reduce the number of laboratory animals and the time-consuming and costly experiments associated with rare cell-type purification.
Collapse
Affiliation(s)
- Katharina Schönberger
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), 79108 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79085 Freiburg, Germany
| | - Michael Mitterer
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Katharina Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), 79108 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79085 Freiburg, Germany
| | - Manuel Stecher
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79085 Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-MCB), 79108 Freiburg, Germany
| | - Sebastian Hobitz
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Dominik Schain-Zota
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Konrad Schuldes
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | | | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| |
Collapse
|
4
|
LIN28A enhances regenerative capacity of human somatic tissue stem cells via metabolic and mitochondrial reprogramming. Cell Death Differ 2022; 29:540-555. [PMID: 34556809 PMCID: PMC8901931 DOI: 10.1038/s41418-021-00873-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Developing methods to improve the regenerative capacity of somatic stem cells (SSCs) is a major challenge in regenerative medicine. Here, we propose the forced expression of LIN28A as a method to modulate cellular metabolism, which in turn enhances self-renewal, differentiation capacities, and engraftment after transplantation of various human SSCs. Mechanistically, in undifferentiated/proliferating SSCs, LIN28A induced metabolic reprogramming from oxidative phosphorylation (OxPhos) to glycolysis by activating PDK1-mediated glycolysis-TCA/OxPhos uncoupling. Mitochondria were also reprogrammed into healthy/fused mitochondria with improved functional capacity. The reprogramming allows SSCs to undergo cell proliferation more extensively with low levels of oxidative and mitochondrial stress. When the PDK1-mediated uncoupling was untethered upon differentiation, LIN28A-SSCs differentiated more efficiently with an increase of OxPhos by utilizing the reprogrammed mitochondria. This study provides mechanistic and practical approaches of utilizing LIN28A and metabolic reprogramming in order to improve SSCs utility in regenerative medicine.
Collapse
|
5
|
Bispo DSC, Jesus CSH, Marques IMC, Romek KM, Oliveira MB, Mano JF, Gil AM. Metabolomic Applications in Stem Cell Research: a Review. Stem Cell Rev Rep 2021; 17:2003-2024. [PMID: 34131883 DOI: 10.1007/s12015-021-10193-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
This review describes the use of metabolomics to study stem cell (SC) characteristics and function, excluding SCs in cancer research, suited to a fully dedicated text. The interest in employing metabolomics in SC research has consistently grown and emphasis is, here, given to developments reported in the past five years. This text informs on the existing methodologies and their complementarity regarding the information provided, comprising untargeted/targeted approaches, which couple mass spectrometry or nuclear magnetic resonance spectroscopy with multivariate analysis (and, in some cases, pathway analysis and integration with other omics), and more specific analytical approaches, namely isotope tracing to highlight particular metabolic pathways, or in tandem microscopic strategies to pinpoint characteristics within a single cell. The bulk of this review covers the existing applications in various aspects of mesenchymal SC behavior, followed by pluripotent and neural SCs, with a few reports addressing other SC types. Some of the central ideas investigated comprise the metabolic/biological impacts of different tissue/donor sources and differentiation conditions, including the importance of considering 3D culture environments, mechanical cues and/or media enrichment to guide differentiation into specific lineages. Metabolomic analysis has considered cell endometabolomes and exometabolomes (fingerprinting and footprinting, respectively), having measured both lipid species and polar metabolites involved in a variety of metabolic pathways. This review clearly demonstrates the current enticing promise of metabolomics in significantly contributing towards a deeper knowledge on SC behavior, and the discovery of new biomarkers of SC function with potential translation to in vivo clinical practice.
Collapse
Affiliation(s)
- Daniela S C Bispo
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - Catarina S H Jesus
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - Inês M C Marques
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - Katarzyna M Romek
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal
| | - Ana M Gil
- Department of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
6
|
Ruud KF, Hiscox WC, Yu I, Chen RK, Li W. Distinct phenotypes of cancer cells on tissue matrix gel. Breast Cancer Res 2020; 22:82. [PMID: 32736579 PMCID: PMC7395363 DOI: 10.1186/s13058-020-01321-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/23/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer cells invading the connective tissues outside the mammary lobule or duct immerse in a reservoir of extracellular matrix (ECM) that is structurally and biochemically distinct from that of their site of origin. The ECM is a spatial network of matrix proteins, which not only provide physical support but also serve as bioactive ligands to the cells. It becomes evident that the dimensional, mechanical, structural, and biochemical properties of ECM are all essential mediators of many cellular functions. To better understand breast cancer development and cancer cell biology in native tissue environment, various tissue-mimicking culture models such as hydrogel have been developed. Collagen I (Col I) and Matrigel are the most common hydrogels used in cancer research and have opened opportunities for addressing biological questions beyond the two-dimensional (2D) cell cultures. Yet, it remains unclear whether these broadly used hydrogels can recapitulate the environmental properties of tissue ECM, and whether breast cancer cells grown on CoI I or Matrigel display similar phenotypes as they would on their native ECM. METHODS We investigated mammary epithelial cell phenotypes and metabolic profiles on animal breast ECM-derived tissue matrix gel (TMG), Col I, and Matrigel. Atomic force microscopy (AFM), fluorescence microscopy, acini formation assay, differentiation experiments, spatial migration/invasion assays, proliferation assay, and nuclear magnetic resonance (NMR) spectroscopy were used to examine biological phenotypes and metabolic changes. Student's t test was applied for statistical analyses. RESULTS Our data showed that under a similar physiological stiffness, the three types of hydrogels exhibited distinct microstructures. Breast cancer cells grown on TMG displayed quite different morphologies, surface receptor expression, differentiation status, migration and invasion, and metabolic profiles compared to those cultured on Col I and Matrigel. Depleting lactate produced by glycolytic metabolism of cancer cells abolished the cell proliferation promoted by the non-tissue-specific hydrogel. CONCLUSION The full ECM protein-based hydrogel system may serve as a biologically relevant model system to study tissue- and disease-specific pathological questions. This work provides insights into tissue matrix regulation of cancer cell biomarker expression and identification of novel therapeutic targets for the treatment of human cancers based on tissue-specific disease modeling.
Collapse
Affiliation(s)
- Kelsey F Ruud
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - William C Hiscox
- Center for NMR Spectroscopy, Washington State University, Pullman, WA, 99164, USA
| | - Ilhan Yu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Roland K Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
7
|
Araújo R, Carneiro TJ, Marinho P, da Costa MM, Roque A, da Cruz E Silva OAB, Fernandes MH, Vilarinho PM, Gil AM. NMR metabolomics to study the metabolic response of human osteoblasts to non-poled and poled poly (L-lactic) acid. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2019; 57:919-933. [PMID: 31058384 DOI: 10.1002/mrc.4883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 06/09/2023]
Abstract
Untargeted nuclear magnetic resonance (NMR) metabolomics was employed, for the first time to our knowledge, to characterize the metabolome of human osteoblast (HOb) cells and extracts in the presence of non-poled or negatively poled poly-L-lactic acid (PLLA). The metabolic response of these cells to this polymer, extensively used in bone regeneration strategies, may potentially translate into useful markers indicative of in vivo biomaterial performance. We present preliminary results of multivariate and univariate analysis of NMR spectra, which have shown the complementarity of lysed cells and extracts in terms of information on cell metabolome, and unveil that, irrespective of poling state, PLLA-grown cells seem to experience enhanced oxidative stress and activated energy metabolism, at the cost of storage lipids and glucose. Possible changes in protein and nucleic acid metabolisms were also suggested, as well as enhanced membrane biosynthesis. Therefore, the presence of PLLA seems to trigger cell catabolism and anti-oxidative protective mechanisms in HOb cells, while directing them towards cellular growth. This was not sufficient, however, to lead to a visible cell proliferation enhancement in the presence of PLLA, although a qualitative tendency for negatively poled PLLA to be more effective in sustaining cell growth than non-poled PLLA was suggested. These preliminary results indicate the potential of NMR metabolomics in enlightening cell metabolism in response to biomaterials and their properties, justifying further studies of the fine effects of poled PLLA on these and other cells of significance in tissue regeneration strategies.
Collapse
Affiliation(s)
- Rita Araújo
- Department of Chemistry and CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Tatiana J Carneiro
- Department of Chemistry and CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Paula Marinho
- Department of Chemistry and CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Marisa Maltez da Costa
- Department of Chemistry and CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
- Department of Materials and Ceramic Engineering, CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Ana Roque
- Department of Medical Sciences, iBIMED-Institute for Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, iBIMED-Institute for Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Maria Helena Fernandes
- Department of Materials and Ceramic Engineering, CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Paula M Vilarinho
- Department of Materials and Ceramic Engineering, CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Aveiro, Portugal
| |
Collapse
|
8
|
Rijal G, Li W. Native-mimicking in vitro microenvironment: an elusive and seductive future for tumor modeling and tissue engineering. J Biol Eng 2018; 12:20. [PMID: 30220913 PMCID: PMC6136168 DOI: 10.1186/s13036-018-0114-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Human connective tissues are complex physiological microenvironments favorable for optimal survival, function, growth, proliferation, differentiation, migration, and death of tissue cells. Mimicking native tissue microenvironment using various three-dimensional (3D) tissue culture systems in vitro has been explored for decades, with great advances being achieved recently at material, design and application levels. These achievements are based on improved understandings about the functionalities of various tissue cells, the biocompatibility and biodegradability of scaffolding materials, the biologically functional factors within native tissues, and the pathophysiological conditions of native tissue microenvironments. Here we discuss these continuously evolving physical aspects of tissue microenvironment important for human disease modeling, with a focus on tumors, as well as for tissue repair and regeneration. The combined information about human tissue spaces reflects the necessities of considerations when configuring spatial microenvironments in vitro with native fidelity to culture cells and regenerate tissues that are beyond the formats of 2D and 3D cultures. It is important to associate tissue-specific cells with specific tissues and microenvironments therein for a better understanding of human biology and disease conditions and for the development of novel approaches to treat human diseases.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| |
Collapse
|
9
|
Nishii K, Brodin E, Renshaw T, Weesner R, Moran E, Soker S, Sparks JL. Shear stress upregulates regeneration-related immediate early genes in liver progenitors in 3D ECM-like microenvironments. J Cell Physiol 2017; 233:4272-4281. [PMID: 29052842 DOI: 10.1002/jcp.26246] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022]
Abstract
The role of fluid stresses in activating the hepatic stem/progenitor cell regenerative response is not well understood. This study hypothesized that immediate early genes (IEGs) with known links to liver regeneration will be upregulated in liver progenitor cells (LPCs) exposed to in vitro shear stresses on the order of those produced from elevated interstitial flow after partial hepatectomy. The objectives were: (1) to develop a shear flow chamber for application of fluid stress to LPCs in 3D culture; and (2) to determine the effects of fluid stress on IEG expression in LPCs. Two hours of shear stress exposure at ∼4 dyn/cm2 was applied to LPCs embedded individually or as 3D spheroids within a hyaluronic acid/collagen I hydrogel. Results were compared against static controls. Quantitative reverse transcriptase polymerase chain reaction was used to evaluate the effect of experimental treatments on gene expression. Twenty-nine genes were analyzed, including IEGs and other genes linked to liver regeneration. Four IEGs (CFOS, IP10, MKP1, ALB) and three other regeneration-related genes (WNT, VEGF, EpCAM) were significantly upregulated in LPCs in response to fluid mechanical stress. LPCs maintained an early to intermediate stage of differentiation in spheroid culture in the absence of the hydrogel, and addition of the gel initiated cholangiocyte differentiation programs which were abrogated by the onset of flow. Collectively the flow-upregulated genes fit the pattern of an LPC-mediated proliferative/regenerative response. These results suggest that fluid stresses are potentially important regulators of the LPC-mediated regeneration response in liver.
Collapse
Affiliation(s)
- Kenichiro Nishii
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio
| | - Erik Brodin
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio
| | - Taylor Renshaw
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio
| | - Rachael Weesner
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio
| | - Emma Moran
- Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina
| | - Jessica L Sparks
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio
| |
Collapse
|
10
|
"Nutrient-sensing" and self-renewal: O-GlcNAc in a new role. J Bioenerg Biomembr 2017; 50:205-211. [PMID: 29204729 DOI: 10.1007/s10863-017-9735-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022]
Abstract
Whether embryonic, hematopoietic or cancer stem cells, this metabolic reprogramming is dependent on the nutrient-status and bioenergetic pathways that is influenced by the micro-environmental niches like hypoxia. Thus, the microenvironment plays a vital role in determining the stem cell fate by inducing metabolic reprogramming. Under the influence of the microenvironment, like hypoxia, the stem cells have increased glucose and glutamine uptake which result in activation of hexosamine biosynthesis pathway (HBP) and increased O-GlcNAc Transferase (OGT). The current review is focused on understanding how HBP, a nutrient-sensing pathway (that leads to increased OGT activity) is instrumental in regulating self-renewal not only in embryonic and hematopoietic stem cells (ESC/HSC) but also in cancer stem cells.
Collapse
|
11
|
Beckervordersandforth R. Mitochondrial Metabolism-Mediated Regulation of Adult Neurogenesis. Brain Plast 2017; 3:73-87. [PMID: 29765861 PMCID: PMC5928529 DOI: 10.3233/bpl-170044] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The life-long generation of new neurons from radial glia-like neural stem cells (NSCs) is achieved through a stereotypic developmental sequence that requires precise regulatory mechanisms to prevent exhaustion or uncontrolled growth of the stem cell pool. Cellular metabolism is the new kid on the block of adult neurogenesis research and the identity of stage-specific metabolic programs and their impact on neurogenesis turns out to be an emerging research topic in the field. Mitochondrial metabolism is best known for energy production but it contains a great deal more. Mitochondria are key players in a variety of cellular processes including ATP synthesis through functional coupling of the electron transport chain and oxidative phosphorylation, recycling of hydrogen carriers, biosynthesis of cellular building blocks, and generation of reactive oxygen species that can modulate signaling pathways in a redox-dependent fashion. In this review, I will discuss recent findings describing stage-specific modulations of mitochondrial metabolism within the adult NSC lineage, emphasizing its importance for NSC self-renewal, proliferation of neural stem and progenitor cells (NSPCs), cell fate decisions, and differentiation and maturation of newborn neurons. I will furthermore summarize the important role of mitochondrial dysfunction in tissue regeneration and ageing, suggesting it as a potential therapeutic target for regenerative medicine practice.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
12
|
Surrati A, Linforth R, Fisk ID, Sottile V, Kim DH. Non-destructive characterisation of mesenchymal stem cell differentiation using LC-MS-based metabolite footprinting. Analyst 2016; 141:3776-87. [PMID: 27102615 DOI: 10.1039/c6an00170j] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bone regeneration is a complex biological process where major cellular changes take place to support the osteogenic differentiation of mesenchymal bone progenitors. To characterise these biological changes and better understand the pathways regulating the formation of mature bone cells, the metabolic profile of mesenchymal stem cell (MSC) differentiation in vitro has been assessed non-invasively during osteogenic (OS) treatment using a footprinting technique. Liquid chromatography (LC)-mass spectrometry (MS)-based metabolite profiling of the culture medium was carried out in parallel to mineral deposition and alkaline phosphatase activity which are two hallmarks of osteogenesis in vitro. Metabolic profiles of spent culture media with a combination of univariate and multivariate analyses investigated concentration changes of extracellular metabolites and nutrients linked to the presence of MSCs in culture media. This non-invasive LC-MS-based analytical approach revealed significant metabolic changes between the media from control and OS-treated cells showing distinct effects of MSC differentiation on the environmental footprint of the cells in different conditions (control vs. OS treatment). A subset of compounds was directly linked to the osteogenic time-course of differentiation, and represent interesting metabolite candidates as non-invasive biomarkers for characterising the differentiation of MSCs in a culture medium.
Collapse
Affiliation(s)
- Amal Surrati
- Wolfson Centre for Stem Cells, Tissue, Engineering and Modelling (STEM), School of Medicine, The University of Nottingham, CBS Building - University Park, Nottingham NG7 2RD, UK.
| | | | | | | | | |
Collapse
|
13
|
Folmes CDL, Terzic A. Energy metabolism in the acquisition and maintenance of stemness. Semin Cell Dev Biol 2016; 52:68-75. [PMID: 26868758 DOI: 10.1016/j.semcdb.2016.02.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 12/17/2022]
Abstract
Energy metabolism is traditionally considered a reactive homeostatic system addressing stage-specific cellular energy needs. There is however growing appreciation of metabolic pathways in the active control of vital cell functions. Case in point, the stem cell lifecycle--from maintenance and acquisition of stemness to lineage commitment and specification--is increasingly recognized as a metabolism-dependent process. Indeed, metabolic reprogramming is an early contributor to the orchestrated departure from or reacquisition of stemness. Recent advances in metabolomics have helped decipher the identity and dynamics of metabolic fluxes implicated in fueling cell fate choices by regulating the epigenetic and transcriptional identity of a cell. Metabolic cues, internal and/or external to the stem cell niche, facilitate progenitor pool restitution, long-term tissue renewal or ensure adoption of cytoprotective behavior. Convergence of energy metabolism with stem cell fate regulation opens a new avenue in understanding primordial developmental biology principles with future applications in regenerative medicine practice.
Collapse
Affiliation(s)
| | - Andre Terzic
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Ji J, Zheng X, Forgues M, Yamashita T, Wauthier EL, Reid LM, Wen X, Song Y, Wei JS, Khan J, Thorgeirsson SS, Wang XW. Identification of microRNAs specific for epithelial cell adhesion molecule-positive tumor cells in hepatocellular carcinoma. Hepatology 2015; 62:829-40. [PMID: 25953724 PMCID: PMC4549211 DOI: 10.1002/hep.27886] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/04/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED Therapies that target cancer stem cells (CSCs) hold promise in eliminating cancer burden. However, normal stem cells are likely to be targeted owing to their similarities to CSCs. It is established that epithelial cell adhesion molecule (EpCAM) is a biomarker for normal hepatic stem cells (HpSCs), and EpCAM(+) AFP(+) hepatocellular carcinoma (HCC) cells have enriched hepatic CSCs. We sought to determine whether specific microRNAs (miRNAs) exist in hepatic CSCs that are not expressed in normal HpSCs. We performed a pair-wise comparison of the miRNA transcriptome of EpCAM(+) and corresponding EpCAM(-) cells isolated from two primary HCC specimens, as well as from two fetal livers and three healthy adult liver donors by small RNA deep sequencing. We found that miR-150, miR-155, and miR-223 were preferentially highly expressed in EpCAM(+) HCC cells, which was further validated. Their gene surrogates, identified using miRNA and messenger RNA profiling in a cohort of 292 HCC patients, were associated with patient prognosis. We further demonstrated that miR-155 was highly expressed in EpCAM(+) HCC cells, compared to corresponding EpCAM(-) HCC cells, fetal livers with enriched normal hepatic progenitors, and normal adult livers with enriched mature hepatocytes. Suppressing miR-155 resulted in a decreased EpCAM(+) fraction in HCC cells and reduced HCC cell colony formation, migration, and invasion in vitro. The reduced levels of identified miR-155 targets predicted the shortened overall survival and time to recurrence of HCC patients. CONCLUSION miR-155 is highly elevated in EpCAM(+) HCC cells and might serve as a molecular target to eradicate the EpCAM(+) CSC population in human HCCs.
Collapse
Affiliation(s)
- Junfang Ji
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China,University of Hawaii Cancer Center, Cancer Biology Program (Ji), Epidemiology Program (Zheng), Honolulu, HI, U.S.A.,Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A.,Corresponding authors: Dr. Xin Wei Wang, National Cancer Institute, 37 Convent Drive, MSC 4258, Bethesda, MD 20892, Tel: +1 301-496-2099, Fax: +1 301-496-0497, ; Dr. Junfang Ji, University of Hawaii Cancer Center, 701 Ilalo Street, Rm 336, Honolulu, HI 96813, Tel: +1 808 441 3492, Fax: +1 808 587 0742, , or Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province, China 310058,
| | - Xin Zheng
- University of Hawaii Cancer Center, Cancer Biology Program (Ji), Epidemiology Program (Zheng), Honolulu, HI, U.S.A
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Eliane L. Wauthier
- Department of Cell Biology and Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, U.S.A
| | - Lola M. Reid
- Department of Cell Biology and Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, U.S.A
| | - Xinyu Wen
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Young Song
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Jun S. Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, U.S.A
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, U.S.A.,Corresponding authors: Dr. Xin Wei Wang, National Cancer Institute, 37 Convent Drive, MSC 4258, Bethesda, MD 20892, Tel: +1 301-496-2099, Fax: +1 301-496-0497, ; Dr. Junfang Ji, University of Hawaii Cancer Center, 701 Ilalo Street, Rm 336, Honolulu, HI 96813, Tel: +1 808 441 3492, Fax: +1 808 587 0742, , or Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province, China 310058,
| |
Collapse
|
15
|
Folmes CDL, Terzic A. Metabolic determinants of embryonic development and stem cell fate. Reprod Fertil Dev 2015; 27:82-8. [PMID: 25472047 DOI: 10.1071/rd14383] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Decoding stem cell metabolism has implicated a tight linkage between energy metabolism and cell fate regulation, a dynamic interplay vital in the execution of developmental and differentiation programs. The inherent plasticity in energy metabolism enables prioritisation of metabolic pathways in support of stage-specific demands. Beyond traditional support of energetic needs, intermediate metabolism may also dictate cell fate choices through regulation of cellular signalling and epigenetic regulation of gene expression. The notion of a 'metabolism-centric' control of stem cell differentiation has been informed by developmental embryogenesis based upon an on-demand paradigm paramount in defining diverse developmental behaviours, from a post-fertilisation nascent zygote to complex organogenesis leading to adequate tissue formation and maturation. Monitored through natural or bioengineered stem cell surrogates, nutrient-responsive metabolites are identified as mediators of cross-talk between metabolic flux, cell signalling and epigenetic regulation charting, collectively, whether a cell will self-renew to maintain progenitor pools, lineage specify to ensure tissue (re)generation or remain quiescent to curb stress damage. Thus, bioenergetics are increasingly recognised as integral in governing stemness and associated organogenic decisions, paving the way for metabolism-defined targets in control of embryology, stem cell biology and tissue regeneration.
Collapse
Affiliation(s)
| | - Andre Terzic
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
16
|
Giri S, Acikgöz A, Bader A. Isolation and Expansion of Hepatic Stem-like Cells from a Healthy Rat Liver and their Efficient Hepatic Differentiation of under Well-defined Vivo Hepatic like Microenvironment in a Multiwell Bioreactor. J Clin Exp Hepatol 2015; 5:107-22. [PMID: 26155038 PMCID: PMC4491607 DOI: 10.1016/j.jceh.2015.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/20/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Currently, undifferentiated cells are found in all tissue and term as local stem cells which are quiescent in nature and less in number under normal healthy conditions but activate upon injury and repair the tissue or organs via automated activating mechanism. Due to very scanty presence of local resident somatic local stem cells in healthy organs, isolation and expansion of these adult stems is an immense challenge for medical research and cell based therapy. Particularly organ like liver, there is an ongoing controversy about existence of liver stem cells. METHODS Herein, Hepatic stem cells population was identified during culture of primary hepatocyte cells upon immediate isolation of primary hepatocyte cells. These liver stem cells has been expanded extensively and differentiated into primary hepatocytes under defined culture conditions in a nanostructured self assembling peptides modular bioreactor that mimic the state of art of liver microenvironment and compared with Matrigel as a positive control. Nanostructured self assembling peptides were used a defined extracellular matrix and Matrigel was used for undefined extracellular matrix. Proliferation of hepatic stem cells was investigated by two strategies. First strategy is to provide high concentration of hepatocyte growth factor (HGF) and second strategy is to evaluate the role of recombinant human erythropoietin (rHuEPO) in presence of trauma/ischemia cytokines (IL-6, TNF-α). Expansion to hepatic differentiation is observed by morphological analysis and was evaluated for the expression of hepatocyte-specific genes using RT-PCR and biochemical methods. RESULTS Hepatocyte-specific genes are well expressed at final stage (day 21) of differentiation period. The differentiated hepatocytes exhibited functional hepatic characteristics such as albumin secretion, urea secretion and cytochrome P450 expression. Additionally, immunofluorescence analysis revealed that hepatic stem cells derived hepatocytes exhibited mature hepatocyte markers (albumin, CK-19, CPY3A1, alpha 1-antitrypsin). Expansion and hepatic differentiation was efficiently in nanostructured self assembling peptides without such batch to batch variation while there was much variation in Matrigel coated bioreactor. In conclusion, the results of the study suggest that the nanostructured self assembling peptides coated bioreactor supports expansion as well as hepatic differentiation of liver stem cells which is superior than Matrigel. CONCLUSION This defined microenvironment conditions in bioreactor module can be useful for research involving bioartificial liver system, stem cell research and engineered liver tissue which could contribute to regenerative cell therapies or drug discovery and development.
Collapse
Key Words
- A1AT, Alpha 1-antitrypsin
- AFP, α-fetoprotein
- CK 7, Cytokeratin 7
- CK-19, Cytokeratin 19
- CPY3A1, Cytochrome P450 3A 1
- EROD, Ethoxyresorufin O-deethylase
- GaIN, D-galactosamine
- HGF, Hepatocyte growth factor
- IL-6, Interleukin 6
- MROD, Methoxyresorufin O-demethylase
- Matrigel
- PROD, Pentoxyresorufin O-depentylase
- TNF-α, Tumor necrosis factor alpha
- Thy1, Thy-1 cell surface antigen
- bioreactor
- defined culture conditions
- hepatic stem cells
- nanostructured self assembling peptides
- rHuEPO, Recombinant human erythropoietin
Collapse
Affiliation(s)
- Shibashish Giri
- Department of Cell Techniques and Applied Stem Cell Biology, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany,Address for correspondence: Shibashish Giri, Department of Cell Techniques and Applied Stem Cell Biology, Center for Biotechnology and Biomedicine, Medical faculty, University of Leipzig, Deutscher Platz 5, D-04103 Leipzig, Germany.
| | - Ali Acikgöz
- Department of Cell Techniques and Applied Stem Cell Biology, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany,Department of Gastroenterology and Hepatology, Klinikum St Georg, Delitzscher Straße, Leipzig, Germany
| | - Augustinus Bader
- Department of Cell Techniques and Applied Stem Cell Biology, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany
| |
Collapse
|
17
|
Arnold JM, Choi WT, Sreekumar A, Maletić-Savatić M. Analytical strategies for studying stem cell metabolism. ACTA ACUST UNITED AC 2015. [PMID: 26213533 DOI: 10.1007/s11515-015-1357-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Owing to their capacity for self-renewal and pluripotency, stem cells possess untold potential for revolutionizing the field of regenerative medicine through the development of novel therapeutic strategies for treating cancer, diabetes, cardiovascular and neurodegenerative diseases. Central to developing these strategies is improving our understanding of biological mechanisms responsible for governing stem cell fate and self-renewal. Increasing attention is being given to the significance of metabolism, through the production of energy and generation of small molecules, as a critical regulator of stem cell functioning. Rapid advances in the field of metabolomics now allow for in-depth profiling of stem cells both in vitro and in vivo, providing a systems perspective on key metabolic and molecular pathways which influence stem cell biology. Understanding the analytical platforms and techniques that are currently used to study stem cell metabolomics, as well as how new insights can be derived from this knowledge, will accelerate new research in the field and improve future efforts to expand our understanding of the interplay between metabolism and stem cell biology.
Collapse
Affiliation(s)
- James M Arnold
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - William T Choi
- Program in Developmental Biology and Medical Scientist Training Program, Baylor College of Medicine; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletić-Savatić
- Program in Developmental Biology and Medical Scientist Training Program, Baylor College of Medicine; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA ; Departments of Pediatrics-Neurology and Neuroscience, and Program in Structural and Computational Biology and Molecular Biophysics Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
18
|
Liu WH, Ren LN, Chen T, You N, Liu LY, Wang T, Yan HT, Luo H, Tang LJ. Unbalanced distribution of materials: the art of giving rise to hepatocytes from liver stem/progenitor cells. J Cell Mol Med 2014; 18:1-14. [PMID: 24286303 PMCID: PMC3916112 DOI: 10.1111/jcmm.12183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/08/2013] [Indexed: 12/12/2022] Open
Abstract
Liver stem/progenitor cells (LSPCs) are able to duplicate themselves and differentiate into each type of cells in the liver, including mature hepatocytes and cholangiocytes. Understanding how to accurately control the hepatic differentiation of LSPCs is a challenge in many fields from preclinical to clinical treatments. This review summarizes the recent advances made to control the hepatic differentiation of LSPCs over the last few decades. The hepatic differentiation of LSPCs is a gradual process consisting of three main steps: initiation, progression and accomplishment. The unbalanced distribution of the affecting materials in each step results in the hepatic maturation of LSPCs. As the innovative and creative works for generating hepatocytes with full functions from LSPCs are gradually accumulated, LSPC therapies will soon be a new choice for treating liver diseases.
Collapse
Affiliation(s)
- Wei-Hui Liu
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Li-Na Ren
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Tao Chen
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Nan You
- Department of General Surgery Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Li-Ye Liu
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Tao Wang
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Hong-Tao Yan
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Hao Luo
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Li-Jun Tang
- General Surgery Center of PLA, Chengdu Military General HospitalChengdu, Sichuan Province, China
| |
Collapse
|
19
|
Folmes CDL, Dzeja PP, Nelson TJ, Terzic A. Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 2013; 11:596-606. [PMID: 23122287 DOI: 10.1016/j.stem.2012.10.002] [Citation(s) in RCA: 514] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Plasticity in energy metabolism allows stem cells to match the divergent demands of self-renewal and lineage specification. Beyond a role in energetic support, new evidence implicates nutrient-responsive metabolites as mediators of crosstalk between metabolic flux, cellular signaling, and epigenetic regulation of cell fate. Stem cell metabolism also offers a potential target for controlling tissue homeostasis and regeneration in aging and disease. In this Perspective, we cover recent progress establishing an emerging relationship between stem cell metabolism and cell fate control.
Collapse
|
20
|
León Z, García-Cañaveras JC, Donato MT, Lahoz A. Mammalian cell metabolomics: experimental design and sample preparation. Electrophoresis 2013; 34:2762-75. [PMID: 23436493 DOI: 10.1002/elps.201200605] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/08/2013] [Accepted: 01/20/2013] [Indexed: 12/19/2022]
Abstract
Metabolomics represents the global assessment of metabolites in a biological sample and reports the closest information to the phenotype of the biological system under study. Mammalian cell metabolomics has emerged as a promising tool with potential applications in many biotechnology and research areas. Metabolomics workflow includes experimental design, sampling, sample processing, metabolite analysis, and data processing. Given their influence on metabolite content and biological interpretation of data, a good experimental design and the appropriate choice of a sample processing method are prerequisites for success in any metabolomic study. The use of mammalian cells in the metabolomics field involves harder sample processing methods, including metabolism quenching and metabolite extraction, as compared to the use of body fluids, although such critical issues are frequently overlooked. This review aims to overview the common experimental procedures used in mammalian cell metabolomics based on mass spectrometry, by placing special emphasis on discussing sample preparation approaches, although other aspects, such as cell metabolomics applications, culture systems, cellular models, analytical platforms, and data analysis, are also briefly covered. This review intends to be a helpful tool to assist researchers in addressing decisions when planning a metabolomics study involving the use of mammalian cells.
Collapse
Affiliation(s)
- Zacarías León
- Unidad Analítica, Instituto de Investigación Sanitaria - Fundación Hospital La Fe, Valencia, Spain
| | | | | | | |
Collapse
|
21
|
Katsuda T, Teratani T, Chowdhury MM, Ochiya T, Sakai Y. Hypoxia efficiently induces differentiation of mouse embryonic stem cells into endodermal and hepatic progenitor cells. Biochem Eng J 2013. [DOI: 10.1016/j.bej.2013.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Turner RA, Wauthier E, Lozoya O, McClelland R, Bowsher JE, Barbier C, Prestwich G, Hsu E, Gerber DA, Reid LM. Successful transplantation of human hepatic stem cells with restricted localization to liver using hyaluronan grafts. Hepatology 2013; 57:775-84. [PMID: 22996260 PMCID: PMC3583296 DOI: 10.1002/hep.26065] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/14/2012] [Indexed: 12/24/2022]
Abstract
Cell therapies are potential alternatives to organ transplantation for liver failure or dysfunction but are compromised by inefficient engraftment, cell dispersal to ectopic sites, and emboli formation. Grafting strategies have been devised for transplantation of human hepatic stem cells (hHpSCs) embedded into a mix of soluble signals and extracellular matrix biomaterials (hyaluronans, type III collagen, laminin) found in stem cell niches. The hHpSCs maintain a stable stem cell phenotype under the graft conditions. The grafts were transplanted into the livers of immunocompromised murine hosts with and without carbon tetrachloride treatment to assess the effects of quiescent versus injured liver conditions. Grafted cells remained localized to the livers, resulting in a larger bolus of engrafted cells in the host livers under quiescent conditions and with potential for more rapid expansion under injured liver conditions. By contrast, transplantation by direct injection or via a vascular route resulted in inefficient engraftment and cell dispersal to ectopic sites. Transplantation by grafting is proposed as a preferred strategy for cell therapies for solid organs such as the liver.
Collapse
Affiliation(s)
- Rachael A. Turner
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Eliane Wauthier
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Oswaldo Lozoya
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Randall McClelland
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - James E. Bowsher
- Department of Biomedical Engineering Duke University School of Medicine, Durham, NC
| | - Claire Barbier
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Glenn Prestwich
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials University of Utah, Salt Lake City, UT
| | - Edward Hsu
- Department of Biomedical Engineering Duke University School of Medicine, Durham, NC
| | - David A. Gerber
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, NC,Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Lola M. Reid
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC,Program in Molecular Biology and Biotechnology, University of North Carolina School of Medicine, Chapel Hill, NC,Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC,Corresponding Author: LM Reid, UNC School of Medicine, Campus Box 7038, Glaxo Building Rms 32-35, Chapel Hill, NC 27599. Phone: 919-966-0347; FAX: 919-6112.
| |
Collapse
|
23
|
Folmes CDL, Martinez-Fernandez A, Faustino RS, Yamada S, Perez-Terzic C, Nelson TJ, Terzic A. Nuclear reprogramming with c-Myc potentiates glycolytic capacity of derived induced pluripotent stem cells. J Cardiovasc Transl Res 2012; 6:10-21. [PMID: 23247633 DOI: 10.1007/s12265-012-9431-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/22/2012] [Indexed: 12/11/2022]
Abstract
Reprogramming strategies influence the differentiation capacity of derived induced pluripotent stem (iPS) cells. Removal of the reprogramming factor c-Myc reduces tumorigenic incidence and increases cardiogenic potential of iPS cells. c-Myc is a regulator of energy metabolism, yet the impact on metabolic reprogramming underlying pluripotent induction is unknown. Here, mitochondrial and metabolic interrogation of iPS cells derived with (4F) and without (3F) c-Myc demonstrated that nuclear reprogramming consistently reverted mitochondria to embryonic-like immature structures. Metabolomic profiling segregated derived iPS cells from the parental somatic source based on the attained pluripotency-associated glycolytic phenotype and discriminated between 3F versus 4F clones based upon glycolytic intermediates. Real-time flux analysis demonstrated a greater glycolytic capacity in 4F iPS cells, in the setting of equivalent oxidative capacity to 3F iPS cells. Thus, inclusion of c-Myc potentiates the pluripotent glycolytic behavior of derived iPS cells, supporting c-Myc-free reprogramming as a strategy to facilitate oxidative metabolism-dependent lineage engagement.
Collapse
Affiliation(s)
- Clifford D L Folmes
- Center for Regenerative Medicine and Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology & Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
LeCluyse EL, Witek RP, Andersen ME, Powers MJ. Organotypic liver culture models: meeting current challenges in toxicity testing. Crit Rev Toxicol 2012; 42:501-48. [PMID: 22582993 PMCID: PMC3423873 DOI: 10.3109/10408444.2012.682115] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 03/26/2012] [Accepted: 03/30/2012] [Indexed: 02/07/2023]
Abstract
Prediction of chemical-induced hepatotoxicity in humans from in vitro data continues to be a significant challenge for the pharmaceutical and chemical industries. Generally, conventional in vitro hepatic model systems (i.e. 2-D static monocultures of primary or immortalized hepatocytes) are limited by their inability to maintain histotypic and phenotypic characteristics over time in culture, including stable expression of clearance and bioactivation pathways, as well as complex adaptive responses to chemical exposure. These systems are less than ideal for longer-term toxicity evaluations and elucidation of key cellular and molecular events involved in primary and secondary adaptation to chemical exposure, or for identification of important mediators of inflammation, proliferation and apoptosis. Progress in implementing a more effective strategy for in vitro-in vivo extrapolation and human risk assessment depends on significant advances in tissue culture technology and increasing their level of biological complexity. This article describes the current and ongoing need for more relevant, organotypic in vitro surrogate systems of human liver and recent efforts to recreate the multicellular architecture and hemodynamic properties of the liver using novel culture platforms. As these systems become more widely used for chemical and drug toxicity testing, there will be a corresponding need to establish standardized testing conditions, endpoint analyses and acceptance criteria. In the future, a balanced approach between sample throughput and biological relevance should provide better in vitro tools that are complementary with animal testing and assist in conducting more predictive human risk assessment.
Collapse
Affiliation(s)
- Edward L LeCluyse
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA.
| | | | | | | |
Collapse
|
25
|
Li J, Xin J, Hao S, Zhang L, Jiang L, Chen D, Xie Q, Xu W, Cao H, Li L. Return of the metabolic trajectory to the original area after human bone marrow mesenchymal stem cell transplantation for the treatment of fulminant hepatic failure. J Proteome Res 2012; 11:3414-3422. [PMID: 22582960 DOI: 10.1021/pr3002639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Our recent study first demonstrated that human bone marrow mesenchymal stem cell (hBMSC) transplantation could prevent death from fulminant hepatic failure (FHF) in pigs. To further clarify the metabolic mechanism of hBMSC transplantation in FHF, the plasma collected from FHF pigs that received transplantation of hBMSCs was examined using metabolic analysis to identify the key molecular markers that regulate recovery. The results showed that obvious metabolic disturbance occurred during FHF, whereas the hBMSC transplantation group showed less severe liver injury. The metabolic trajectory returns to its original state at week 3 following the hBMSC transplantation. In total, the concentration of 26 metabolites, including conjugated bile acids, phosphatidylcholines, lysophosphatidylcholines, fatty acids, amino acid and sphingomyelin, are significantly different between the FHF group and the hBMSC transplantation group. Moreover, the time course of changes in the metabolites corresponded with that of the biochemical and histological analyses. Real-time PCR further confirmed that the gene expression of phospholipase A1, lecithin-cholesterol acyltransferase and lysophosphatidylcholine acyltransferase 1 decreased significantly, whereas that of phospholipase A2 remained stable, which explains the decrease of the phosphatidylcholines and lysophosphatidylcholines. These novel results have revealed a metabolic mechanism for the hBMSC transplantation in FHF, which could lead to the future development of treatment strategies for stem cell therapies.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University , 79 Qingchun Rd., Hangzhou 310003, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Turner RA, Mendel G, Wauthier E, Barbier C, Reid LM. Hyaluronan-supplemented buffers preserve adhesion mechanisms facilitating cryopreservation of human hepatic stem/progenitor cells. Cell Transplant 2012; 21:2257-66. [PMID: 22472355 DOI: 10.3727/096368912x637000] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The supply of human hepatic stem cells (hHpSCs) and other hepatic progenitors has been constrained by the limited availability of liver tissues from surgical resections, the rejected organs from organ donation programs, and the need to use cells immediately. To facilitate accessibility to these precious tissue resources, we have established an effective method for serum-free cryopreservation of the cells, allowing them to be stockpiled and stored for use as an off-the-shelf product for experimental or clinical programs. The method involves use of buffers, some serum-free, designed for cryopreservation and further supplemented with hyaluronans (HA) that preserve adhesion mechanisms facilitating postthaw culturing of the cells and preservation of functions. Multiple cryopreservation buffers were found to yield high viabilities (80-90%) of cells on thawing of the progenitor cells. Serum-free CS10 supplemented with 0.05% hyaluronan proved the most effective, both in terms of viabilities of cells on thawing and in yielding cell attachment and formation of expanding colonies of cells that stably maintain the stem/progenitor cell phenotype. Buffers to which 0.05 or 0.1% HAs were added showed cells postthaw to be phenotypically stable as stem/progenitors, as well as having a high efficiency of attachment and expansion in culture. Success correlated with improved expression of adhesion molecules, particularly CD44, the hyaluronan receptor, E-cadherin, β4 integrin in hHpSCs, and β1 integrins in hepatoblasts. The improved methods in cryopreservation offer more efficient strategies for stem cell banking in both research and potential therapy applications.
Collapse
Affiliation(s)
- Rachael A Turner
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
Nuclear reprogramming with stemness factors enables resetting of somatic differentiated tissue back to the pluripotent ground state. Recent evidence implicates mitochondrial restructuring and bioenergetic plasticity as key components underlying execution of orchestrated dedifferentiation and derivation of induced pluripotent stem cells. Aerobic to anaerobic transition of somatic oxidative energy metabolism into a glycolytic metabotype promotes proficient reprogramming, establishing a novel regulator of acquired stemness. Metabolomic profiling has further identified specific metabolic remodeling traits defining lineage redifferentiation of pluripotent cells. Therefore, mitochondrial biogenesis and energy metabolism comprise a vital axis for biomarker discovery, intimately reflecting the molecular dynamics fundamental for the resetting and redirection of cell fate.
Collapse
Affiliation(s)
- Clifford D L Folmes
- Center for Regenerative Medicine and Marriott Heart Disease Research Program, MN, USA
| | | | | |
Collapse
|
28
|
Cardinale V, Wang Y, Carpino G, Mendel G, Alpini G, Gaudio E, Reid LM, Alvaro D. The biliary tree--a reservoir of multipotent stem cells. Nat Rev Gastroenterol Hepatol 2012; 9:231-40. [PMID: 22371217 DOI: 10.1038/nrgastro.2012.23] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The biliary tree is composed of intrahepatic and extrahepatic bile ducts, lined by mature epithelial cells called cholangiocytes, and contains peribiliary glands deep within the duct walls. Branch points, such as the cystic duct, perihilar and periampullar regions, contain high numbers of these glands. Peribiliary glands contain multipotent stem cells, which self-replicate and can differentiate into hepatocytes, cholangiocytes or pancreatic islets, depending on the microenvironment. Similar cells-presumably committed progenitor cells-are found in the gallbladder (which lacks peribiliary glands). The stem and progenitor cell characteristics indicate a common embryological origin for the liver, biliary tree and pancreas, which has implications for regenerative medicine as well as the pathophysiology and oncogenesis of midgut organs. This Perspectives article describes a hypothetical model of cell lineages starting in the duodenum and extending to the liver and pancreas, and thought to contribute to ongoing organogenesis throughout life.
Collapse
Affiliation(s)
- Vincenzo Cardinale
- Division of Gastroenterology, Department of Medico-Surgical Sciences and Biotechnology, Fondazione Eleonora Lorillard Spencer Cenci, Polo Pontino, Corso della Repubblica 79, 04100 Latina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
McClelland R, Wauthier E, Tallheden T, Reid L, Hsu E. In situ labeling and magnetic resonance imaging of transplanted human hepatic stem cells. Mol Imaging Biol 2011; 13:911-22. [PMID: 20890665 PMCID: PMC3727160 DOI: 10.1007/s11307-010-0422-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The purpose is to address the problem in magnetic resonance imaging (MRI) of contrast agent dilution. PROCEDURES In situ magnetic labeling of cells and MRI were used to assess distribution and growth of human hepatic stem cells (hHpSCs) transplanted into severe combined immunodeficiency (SCID)/non-obese diabetic (NOD) mice. It was done with commercially available magnetic microbeads coupled to an antibody to a surface antigen, epithelial cell adhesion molecule (EpCAM), uniquely expressed in the liver by hepatic progenitors. RESULTS We validated the microbead connection to cells and related MRI data to optical microscopy observations in order to develop a means to quantitatively estimate cell numbers in the aggregates detected. Cell counts of hHpSCs at different times post-transplantation revealed quantifiable evidence of cell engraftment and expansion. CONCLUSIONS This magnetic labeling methodology can be used with any antibody coupled to a magnetic particle to target any surface antigen that distinguishes transplanted cells from host cells, thus facilitating studies that define methods and strategies for clinical cell therapy programs.
Collapse
Affiliation(s)
- Randall McClelland
- Department of Cell and Molecular Physiology, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
| | - Eliane Wauthier
- Department of Cell and Molecular Physiology, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
| | - Tommi Tallheden
- Department of Cell and Molecular Physiology, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
| | - Lola Reid
- Department of Cell and Molecular Physiology, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
- Department of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
- Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC 27599, USA 27599
| | - Edward Hsu
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Durham, NC 27708
| |
Collapse
|
30
|
Lozoya OA, Wauthier E, Turner R, Barbier C, Prestwich GD, Guilak F, Superfine R, Lubkin SR, Reid LM. Regulation of hepatic stem/progenitor phenotype by microenvironment stiffness in hydrogel models of the human liver stem cell niche. Biomaterials 2011; 32:7389-402. [PMID: 21788068 PMCID: PMC3157321 DOI: 10.1016/j.biomaterials.2011.06.042] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/20/2011] [Indexed: 02/09/2023]
Abstract
Human livers have maturational lineages of cells within liver acini, beginning periportally in stem cell niches, the canals of Hering, and ending in polyploid hepatocytes pericentrally and cholangiocytes in bile ducts. Hepatic stem cells (hHpSCs) in vivo are partnered with mesenchymal precursors to endothelia (angioblasts) and stellate cells, and reside in regulated microenvironments, stem cell niches, containing hyaluronans (HA). The in vivo hHpSC niche is modeled in vitro by growing hHpSC in two-dimensional (2D) cultures on plastic. We investigated effects of 3D microenvironments, mimicking the liver's stem cell niche, on these hHpSCs by embedding them in HA-based hydrogels prepared with Kubota's Medium (KM), a serum-free medium tailored for endodermal stem/progenitors. The KM-HA hydrogels mimicked the niches, matched diffusivity of culture medium, exhibited shear thinning and perfect elasticity under mechanical loading, and had predictable stiffness depending on their chemistry. KM-HA hydrogels, which supported cell attachment, survival and expansion of hHpSC colonies, induced transition of hHpSC colonies towards stable heterogeneous populations of hepatic progenitors depending on KM-HA hydrogel stiffness, as shown by both their gene and protein expression profile. These acquired phenotypes did not show morphological evidence of fibrotic responses. In conclusion, this study shows that the mechanical properties of the microenvironment can regulate differentiation in endodermal stem cell populations.
Collapse
Affiliation(s)
- Oswaldo A. Lozoya
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, Lineberger Comprehensive Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, UNC School of Medicine, Chapel Hill, NC
| | - Eliane Wauthier
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, Lineberger Comprehensive Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, UNC School of Medicine, Chapel Hill, NC
| | - Rachael Turner
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, Lineberger Comprehensive Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, UNC School of Medicine, Chapel Hill, NC
| | - Claire Barbier
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, Lineberger Comprehensive Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, UNC School of Medicine, Chapel Hill, NC
| | - Glenn D. Prestwich
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, University of Utah, Salt Lake City, UT
| | - Farshid Guilak
- Departments of Surgery, Biomedical Engineering, Mechanical Engineering and Materials Science, Duke University Medical Center and Pratt School of Engineering, Durham, NC
| | - Richard Superfine
- Department of Physics and Astronomy, UNC College of Arts and Sciences, Chapel Hill, NC
| | - Sharon R. Lubkin
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Mathematics, North Carolina State University, Raleigh, NC
| | - Lola M. Reid
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC and UNC School of Medicine, Chapel Hill, NC
- Department of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, Lineberger Comprehensive Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, UNC School of Medicine, Chapel Hill, NC
| |
Collapse
|
31
|
Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metab 2011; 14:264-71. [PMID: 21803296 PMCID: PMC3156138 DOI: 10.1016/j.cmet.2011.06.011] [Citation(s) in RCA: 776] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 05/01/2011] [Accepted: 06/09/2011] [Indexed: 12/12/2022]
Abstract
The bioenergetics of somatic dedifferentiation into induced pluripotent stem cells remains largely unknown. Here, stemness factor-mediated nuclear reprogramming reverted mitochondrial networks into cristae-poor structures. Metabolomic footprinting and fingerprinting distinguished derived pluripotent progeny from parental fibroblasts according to elevated glucose utilization and production of glycolytic end products. Temporal sampling demonstrated glycolytic gene potentiation prior to induction of pluripotent markers. Functional metamorphosis of somatic oxidative phosphorylation into acquired pluripotent glycolytic metabolism conformed to an embryonic-like archetype. Stimulation of glycolysis promoted, while blockade of glycolytic enzyme activity blunted, reprogramming efficiency. Metaboproteomics resolved upregulated glycolytic enzymes and downregulated electron transport chain complex I subunits underlying cell fate determination. Thus, the energetic infrastructure of somatic cells transitions into a required glycolytic metabotype to fuel induction of pluripotency.
Collapse
|
32
|
Prestwich GD. Hyaluronic acid-based clinical biomaterials derived for cell and molecule delivery in regenerative medicine. J Control Release 2011; 155:193-9. [PMID: 21513749 DOI: 10.1016/j.jconrel.2011.04.007] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 02/01/2023]
Abstract
The development of injectable and biocompatible vehicles for delivery, retention, growth, and differentiation of stem cells is of paramount importance for regenerative medicine. For cell therapy and the development of clinical combination products, we created a hyaluronan (HA)-based synthetic extracellular matrix (sECM) that provides highly reproducible, manufacturable, approvable, and affordable biomaterials. The composition of the sECM can be customized for use with progenitor and mature cell populations obtained from skin, fat, liver, heart, muscle, bone, cartilage, nerves, and other tissues. This overview describes the design criteria for "living" HA derivatives, and the many uses of this in situ crosslinkable HA-based sECM hydrogel for three-dimensional (3-D) culture of cells in vitro and translational use in vivo. Recent advances allow rapid expansion and recovery of cells in 3-D, and the bioprinting of engineered tissue constructs. The uses of HA-derived sECMs for cell and molecule delivery in vivo will be reviewed, including applications in cancer biology and tumor imaging.
Collapse
Affiliation(s)
- Glenn D Prestwich
- Center for Therapeutic Biomaterials and Department of Medicinal Chemistry, University of Utah, 419 Wakara Way #205, Salt Lake City, UT 84108-1257, USA.
| |
Collapse
|
33
|
Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H41-56. [PMID: 21394792 PMCID: PMC3730855 DOI: 10.1002/adma.201003963] [Citation(s) in RCA: 1413] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 01/03/2011] [Indexed: 05/10/2023]
Abstract
Hyaluronic acid (HA), an immunoneutral polysaccharide that is ubiquitous in the human body, is crucial for many cellular and tissue functions and has been in clinical use for over thirty years. When chemically modified, HA can be transformed into many physical forms-viscoelastic solutions, soft or stiff hydrogels, electrospun fibers, non-woven meshes, macroporous and fibrillar sponges, flexible sheets, and nanoparticulate fluids-for use in a range of preclinical and clinical settings. Many of these forms are derived from the chemical crosslinking of pendant reactive groups by addition/condensation chemistry or by radical polymerization. Clinical products for cell therapy and regenerative medicine require crosslinking chemistry that is compatible with the encapsulation of cells and injection into tissues. Moreover, an injectable clinical biomaterial must meet marketing, regulatory, and financial constraints to provide affordable products that can be approved, deployed to the clinic, and used by physicians. Many HA-derived hydrogels meet these criteria, and can deliver cells and therapeutic agents for tissue repair and regeneration. This progress report covers both basic concepts and recent advances in the development of HA-based hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Jason A. Burdick
- Prof. J.A. Burdick, Department of Bioengineering, University of Pennsylvania, 210 S 33th Street, Philadelphia, PA 19104 (USA),
| | - Glenn D. Prestwich
- Prof. G.D. Prestwich, Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, University of Utah, 419 Wakara Way, Suite 205, Salt Lake City, UT 84108 (USA),
| |
Collapse
|
34
|
Abstract
The detailed knowledge of mammalian cell metabolism and its adjustments to different cell properties and perturbations, such as disease and drug exposure, is of enormous value in the deeper understanding of pathological processes and drug mechanisms, as well as in the development of new and improved methods for diagnosis, follow-up of disease progression and treatment response. This review covers recent developments in the use of NMR-based metabonomics to characterize cellular metabolomes and interpret them in terms of metabolic changes taking place in a wide range of situations. The analytical methodology available is briefly presented and the applications developed so far are reviewed. These include differences in cell properties (e.g., drug resistance, cell cycle stage, specific growth conditions and genetic characteristics) and changes induced in response to different perturbations (e.g., disease, drug exposure and irradiation).
Collapse
|
35
|
Turner R, Lozoya O, Wang Y, Cardinale V, Gaudio E, Alpini G, Mendel G, Wauthier E, Barbier C, Alvaro D, Reid LM. Human hepatic stem cell and maturational liver lineage biology. Hepatology 2011; 53:1035-45. [PMID: 21374667 PMCID: PMC3066046 DOI: 10.1002/hep.24157] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Livers are comprised of maturational lineages of cells beginning extrahepatically in the hepato-pancreatic common duct near the duodenum and intrahepatically in zone 1 by the portal triads. The extrahepatic stem cell niches are the peribiliary glands deep within the walls of the bile ducts; those intrahepatically are the canals of Hering in postnatal livers and that derive from ductal plates in fetal livers. Intrahepatically, there are at least eight maturational lineage stages from the stem cells in zone 1 (periportal), through the midacinar region (zone 2), to the most mature cells and apoptotic cells found pericentrally in zone 3. Those found in the biliary tree are still being defined. Parenchymal cells are closely associated with lineages of mesenchymal cells, and their maturation is coordinated. Each lineage stage consists of parenchymal and mesenchymal cell partners distinguishable by their morphology, ploidy, antigens, biochemical traits, gene expression, and ability to divide. They are governed by changes in chromatin (e.g., methylation), gradients of paracrine signals (soluble factors and insoluble extracellular matrix components), mechanical forces, and feedback loop signals derived from late lineage cells. Feedback loop signals, secreted by late lineage stage cells into bile, flow back to the periportal area and regulate the stem cells and other early lineage stage cells in mechanisms dictating the size of the liver mass. Recognition of maturational lineage biology and its regulation by these multiple mechanisms offers new understandings of liver biology, pathologies, and strategies for regenerative medicine and treatment of liver cancers.
Collapse
Affiliation(s)
- Rachael Turner
- University of North Carolina School of Medicine, Department of Cell and Molecular Physiology, Chapel Hill, North Carolina 27599
- University of North Carolina School of Medicine, Department of Biomedical Engineering, Chapel Hill, North Carolina 27599
| | - Oswaldo Lozoya
- University of North Carolina School of Medicine, Department of Biomedical Engineering, Chapel Hill, North Carolina 27599
| | - Yunfang Wang
- University of North Carolina School of Medicine, Department of Cell and Molecular Physiology, Chapel Hill, North Carolina 27599
| | - Vincenzo Cardinale
- Division of Gastroenterology, Department of Clinical Medicine, University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Human Anatomy, University of Rome, Rome, Italy
| | - Gianfranco Alpini
- Division of Research, Central Texas Veterans Health Care System, Department of Medicine, Scott & White Digestive Disease Research Center, Division of Research and Education, Scott & White and Texas A&M Health Science Center College of Medicine, Temple, TX, 76504
| | - Gemma Mendel
- University of North Carolina School of Medicine, Department of Biomedical Engineering, Chapel Hill, North Carolina 27599
| | - Eliane Wauthier
- University of North Carolina School of Medicine, Department of Cell and Molecular Physiology, Chapel Hill, North Carolina 27599
| | - Claire Barbier
- University of North Carolina School of Medicine, Department of Cell and Molecular Physiology, Chapel Hill, North Carolina 27599
| | - Domenico Alvaro
- Division of Gastroenterology, Department of Clinical Medicine, University of Rome, Rome, Italy
| | - Lola M. Reid
- University of North Carolina School of Medicine, Department of Cell and Molecular Physiology, Chapel Hill, North Carolina 27599
- University of North Carolina School of Medicine, Department of Biomedical Engineering, Chapel Hill, North Carolina 27599
- University of North Carolina School of Medicine, Program in Molecular Biology and Biotechnology, Chapel Hill, North Carolina 27599
| |
Collapse
|
36
|
Skardal A, Sarker SF, Crabbé A, Nickerson CA, Prestwich GD. The generation of 3-D tissue models based on hyaluronan hydrogel-coated microcarriers within a rotating wall vessel bioreactor. Biomaterials 2010; 31:8426-35. [PMID: 20692703 DOI: 10.1016/j.biomaterials.2010.07.047] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Accepted: 07/08/2010] [Indexed: 10/19/2022]
Abstract
With the increasing necessity for functional tissue- and organ equivalents in the clinic, the optimization of techniques for the in vitro generation of organotypic structures that closely resemble the native tissue is of paramount importance. The engineering of a variety of highly differentiated tissues has been achieved using the rotating wall vessel (RWV) bioreactor technology, which is an optimized suspension culture allowing cells to grow in three-dimensions (3-D). However, certain cell types require the use of scaffolds, such as collagen-coated microcarrier beads, for optimal growth and differentiation in the RWV. Removal of the 3-D structures from the microcarriers involves enzymatic treatment, which disrupts the delicate 3-D architecture and makes it inapplicable for potential implantation. Therefore, we designed a microcarrier bead coated with a synthetic extracellular matrix (ECM) composed of a disulfide-crosslinked hyaluronan and gelatin hydrogel for 3-D tissue engineering, that allows for enzyme-free cell detachment under mild reductive conditions (i.e. by a thiol-disulfide exchange reaction). The ECM-coated beads (ECB) served as scaffold to culture human intestinal epithelial cells (Int-407) in the RWV, which formed viable multi-layered cell aggregates and expressed epithelial differentiation markers. The cell aggregates remained viable following dissociation from the microcarriers, and could be returned to the RWV bioreactor for further culturing into bead-free tissue assemblies. The developed ECBs thus offer the potential to generate scaffold-free 3-D tissue assemblies, which could further be explored for tissue replacement and remodeling.
Collapse
Affiliation(s)
- Aleksander Skardal
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84108-1257, USA
| | | | | | | | | |
Collapse
|
37
|
Nuclear magnetic resonance detects phosphoinositide 3-kinase/Akt-independent traits common to pluripotent murine embryonic stem cells and their malignant counterparts. Neoplasia 2010; 11:1301-8. [PMID: 20019838 DOI: 10.1593/neo.09850] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 08/10/2009] [Accepted: 08/10/2009] [Indexed: 12/30/2022] Open
Abstract
Pluripotent embryonic stem (ES) cells, a potential source of somatic precursors for cell therapies, cause tumors after transplantation. Studies of mammalian carcinogenesis using nuclear magnetic resonance (NMR) spectroscopy have revealed changes in the choline region, particularly increased phosphocholine (PCho) content. High PCho levels in murine ES (mES) cells have recently been attributed to cell pluripotency. The phosphoinositide 3-kinase (PI3K)/Akt pathway has been implicated in tumor-like properties of mES cells. This study aimed to examine a potential link between the metabolic profile associated with choline metabolism of pluripotent mES cells and PI3K/Akt signaling. We used mES (ES-D3) and murine embryonal carcinoma cells (EC-F9) and compared the metabolic profiles of 1) pluripotent mES (ESD0), 2) differentiated mES (ESD14), and 3) pluripotent F9 cells. Involvement of the PI3K/Akt pathway was assessed using LY294002, a selective PI3K inhibitor. Metabolic profiles were characterized in the extracted polar fraction by (1)H NMR spectroscopy. Similarities were found between the levels of choline phospholipid metabolites (PCho/total choline and PCho/glycerophosphocholine [GPCho]) in ESD0 and F9 cell spectra and a greater-than five-fold decrease of the PCho/GPCho ratio associated with mES cell differentiation. LY294002 caused no significant change in relative PCho levels but led to a greater-than two-fold increase in PCho/GPCho ratios. These results suggest that the PCho/GPCho ratio is a metabolic trait shared by pluripotent and malignant cells and that PI3K does not underlie its development. It is likely that the signature identified here in a mouse model may be relevant for safe therapeutic applications of human ES cells.
Collapse
|
38
|
Markert CD, Atala A, Cann JK, Christ G, Furth M, Ambrosio F, Childers MK. Mesenchymal stem cells: emerging therapy for Duchenne muscular dystrophy. PM R 2009; 1:547-59. [PMID: 19627945 DOI: 10.1016/j.pmrj.2009.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 02/20/2009] [Accepted: 02/25/2009] [Indexed: 12/31/2022]
Abstract
Multipotent cells that can give rise to bone, cartilage, fat, connective tissue, and skeletal and cardiac muscle are termed mesenchymal stem cells. These cells were first identified in the bone marrow, distinct from blood-forming stem cells. Based on the embryologic derivation, availability, and various pro-regenerative characteristics, research exploring their use in cell therapy shows great promise for patients with degenerative muscle diseases and a number of other conditions. In this review, the authors explore the potential for mesenchymal stem cell therapy in the emerging field of regenerative medicine with a focus on treatment for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Chad D Markert
- Department of Neurology, School of Medicine, and Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
McClelland R, Wauthier E, Zhang L, Melhem A, Schmelzer E, Barbier C, Reid LM. Ex vivo conditions for self-replication of human hepatic stem cells. Tissue Eng Part C Methods 2008; 14:341-351. [PMID: 18844603 PMCID: PMC2913780 DOI: 10.1089/ten.tec.2008.0073] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 07/01/2008] [Indexed: 12/16/2022] Open
Abstract
Human hepatic stem cells (hHpSCs), identifiable by a unique antigenic profile, have been isolated from human livers and established ex vivo under expansion conditions permissive for self-replication. The conditions consist of a substratum of type III collagen, ideally on Transwell inserts, and Kubota's medium, a serum-free medium developed for hepatic progenitors. Under these conditions the cells demonstrated a doubling time of approximately 24 h, generating at least a 16-fold increase in cell number within 7-10 days; were stable at confluence for up to 2 weeks; could be passaged, if on type III collagen, to initiate colonies that went through log-phase growth and saturation density kinetics; and expressed telomerase, indicative of regenerative capacity. The hHpSC colonies remained morphologically and phenotypically stable throughout expressing epithelial cell adhesion molecule, neural cell adhesion molecule, albumin, cytokeratins 8, 18, and 19, but not alpha-fetoprotein, or intercellular adhesion molecule-1 (ICAM-1). Those maintained under self-replication conditions for more than a month were transplanted and found to engraft in the livers of SCID/nod mice yielding human liver tissue expressing adult liver-specific proteins. The conditions for self-replication should offer ideal culture conditions for generating large numbers of hHpSCs for use in commercial and clinical programs.
Collapse
Affiliation(s)
- Randall McClelland
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
- Current: Admet Technologies, Durham, North Carolina
| | - Eliane Wauthier
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
| | - Lili Zhang
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
- Current: Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR of China
| | - Alaa Melhem
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
- Current: Department of Gastroenterology and Hepatology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eva Schmelzer
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
- Current: Department of Surgery, University of Pittsburgh, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Claire Barbier
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
| | - Lola M. Reid
- Departments of Cell and Molecular Physiology and Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, North Carolina
- Department of Biomedical Engineering, Cancer Center and Center for Gastrointestinal and Biliary Disease Biology (CGIBD), UNC School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
40
|
Zhang L, Theise N, Chua M, Reid LM. The stem cell niche of human livers: symmetry between development and regeneration. Hepatology 2008; 48:1598-1607. [PMID: 18972441 DOI: 10.1002/hep.22516] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human livers contain two pluripotent progenitors: hepatic stem cells and hepatoblasts. The hepatic stem cells uniquely express the combination of epithelial cell adhesion molecule (EpCAM), neural cell adhesion molecule (NCAM), cytokeratin (CK) 19, albumin +/-, and are negative for alpha-fetoprotein (AFP). They are precursors to hepatoblasts, which differ from hepatic stem cells in size, morphology, and in expressing the combination of EpCAM, intercellular cell adhesion molecule (ICAM-1), CK19, albumin++, and AFP++. The hepatic stem cells are located in vivo in stem cell niches: the ductal plates in fetal and neonatal livers and canals of Hering in pediatric and adult livers. The hepatoblasts are contiguous to the niches, decline in numbers with age, wax and wane in numbers with injury responses, and are proposed to be the liver's transit-amplifying cells. In adult livers, intermediates between hepatic stem cells and hepatoblasts and between hepatoblasts and adult parenchyma are observed. Amplification of one or both pluripotent cell subpopulations can occur in diseases; for example, hepatic stem cell amplification occurs in mild forms of liver failure, and hepatoblast amplification occurs in forms of cirrhosis. Liver is, therefore, similar to other tissues in that regenerative processes in postnatal tissues parallel those occurring in development and involve populations of stem cells and progenitor cells that can be identified by anatomic, antigenic, and biochemical profiles.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
41
|
Zhang J, Skardal A, Prestwich GD. Engineered extracellular matrices with cleavable crosslinkers for cell expansion and easy cell recovery. Biomaterials 2008; 29:4521-31. [PMID: 18768219 DOI: 10.1016/j.biomaterials.2008.08.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 07/30/2008] [Accepted: 08/04/2008] [Indexed: 10/21/2022]
Abstract
An unmet need for expansion of primary cells and progenitor cells in three dimensions (3-D) is a synthetic mimic of the extracellular matrix (ECM) with user-controllable composition that would permit rapid recovery of viable cells under mild, non-enzymatic conditions. Three block copolymers based on disulfide-containing polyethylene glycol diacrylate crosslinkers were synthesized, and were used to crosslink thiol-modified hyaluronan and gelatin macromonomers in the presence of cells. The triblock PEGSSDA contained a single disulfide-containing block, the pentablock PEG(SS)(2)DA contained two disulfide blocks, and the heptablock PEG(SS)(3)DA contained three disulfide blocks. For each hydrogel composition, four cell types were encapsulated in 3-D, and growth and proliferation were evaluated. Murine NIH 3T3 fibroblasts, human HepG2 C3A hepatocytes, human bone marrow-derived mesenchymal stem cells (MSCs), and human umbilical vein endothelial cells (HUVECs) all showed excellent viability and growth during expansion in 3-D in the three disulfide block copolymer crosslinkers. After cell expansion, the hydrogels were dissociated using the thiol-disulfide exchange reaction in the presence of N-acetyl-cysteine or glutathione, which dissolved the hydrogel network. After dissolution, cells were recovered in high yield and with high viability by gentle centrifugation.
Collapse
Affiliation(s)
- Jianxing Zhang
- Center for Therapeutic Biomaterials, Department of Medicinal Chemistry, The University of Utah, 419 Wakara Way Suite 205, Salt Lake City, UT 84108-1257, USA
| | | | | |
Collapse
|