1
|
Choi G, Tang Z, Guan W. Microfluidic high-throughput single-cell mechanotyping: Devices and
applications. NANOTECHNOLOGY AND PRECISION ENGINEERING 2021. [DOI: 10.1063/10.0006042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Gihoon Choi
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| | - Zifan Tang
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| | - Weihua Guan
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| |
Collapse
|
2
|
Samal P, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Grow with the Flow: When Morphogenesis Meets Microfluidics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805764. [PMID: 30767289 DOI: 10.1002/adma.201805764] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Developmental biology has advanced the understanding of the intricate and dynamic processes involved in the formation of an organism from a single cell. However, many gaps remain in the knowledge of embryonic development, especially regarding tissue morphogenesis. A possible approach to mimic such phenomena uses pluripotent stem cells in in vitro morphogenetic models. Herein, these systems are summarized with emphasis on the ability to better manipulate and control cellular interfaces with either liquid or solid materials using microengineered tools, which is critical for attaining deeper insights into pattern formation and stem cell differentiation during organogenesis. The role of conventional and customized cell-culture systems in supporting important advances in the field of morphogenesis is discussed, and the fascinating role that material sciences and microengineering currently play and are expected to play in the future is highlighted. In conclusion, it is proffered that continued microfluidics innovations when applied to morphogenesis promise to provide important insights to advance many multidisciplinary fields, including regenerative medicine.
Collapse
Affiliation(s)
- Pinak Samal
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
3
|
Banerjee H, Roy B, Chaudhury K, Srinivasan B, Chakraborty S, Ren H. Frequency-induced morphology alterations in microconfined biological cells. Med Biol Eng Comput 2018; 57:819-835. [PMID: 30415434 DOI: 10.1007/s11517-018-1908-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 09/29/2018] [Indexed: 01/09/2023]
Abstract
Low-intensity therapeutic ultrasound has demonstrated an impetus in bone signaling and tissue healing for decades now. Though this technology is clinically well proven, still there are breaches in studies to understand the fundamental principle of how osteoblast tissue regenerates physiologically at the cellular level with ultrasound interaction as a form of acoustic wave stimuli. Through this article, we illustrate an analysis for cytomechanical changes of cell membrane periphery as a basic first physical principle for facilitating late downstream biochemical pathways. With the help of in situ single-cell direct analysis in a microfluidic confinement, we demonstrate that alteration of low-intensity pulse ultrasound (LIPUS) frequency would physically perturb cell membrane and establish inherent cell oscillation. We experimentally demonstrate here that, at LIPUS resonance near 1.7 MHz (during 1-3 MHz alteration), cell membrane area would expand to 6.85 ± 0.7% during ultrasound exposure while it contracts 44.68 ± 0.8% in post actuation. Conversely, cell cross-sectional area change (%) from its previous morphology during and after switching off LIPUS was reversibly different before and after resonance. For instance, at 1.5 MHz, LIPUS exposure produced 1.44 ± 0.5% expansion while in contrast 2 MHz instigates 1.6 ± 0.3% contraction. We conclude that alteration of LIPUS frequency from 1-3 MHz keeping other ultrasound parameters like exposure time, pulse repetition frequency (PRF), etc., constant, if applied to a microconfined biological single living cell, would perturb physical structure reversibly based on the system resonance during and post exposure ultrasound pulsing. We envision, in the near future, our results would constitute the foundation of mechanistic effects of low-intensity therapeutic ultrasound and its allied potential in medical applications. Graphical Abstract Frequency Dependent Characterization of Area Strain in Cell Membrane by Microfluidic Based Single Cell Analysis.
Collapse
Affiliation(s)
- Hritwick Banerjee
- Department of Electrical Engineering, Indian Institute of Technology Gandhinagar, Village Palaj Simkheda, Gandhinagar, Gujarat, 382355, India. .,Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India. .,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore, 117575, Singapore. .,Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, #05-COR, Singapore, 117456, Singapore.
| | - Bibhas Roy
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.,Mechanobiology Institute, National University of Singapore, T-Lab, #10-01 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Kaustav Chaudhury
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.,National Institute of Technology Rourkela, Odisha, 769008, India
| | - Babji Srinivasan
- Department of Electrical Engineering, Indian Institute of Technology Gandhinagar, Village Palaj Simkheda, Gandhinagar, Gujarat, 382355, India.,Department of Chemical Engineering, Indian Institute of Technology Gandhinagar, Village Palaj Simkheda, Gandhinagar, Gujarat, 382355, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.,School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Hongliang Ren
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore, 117575, Singapore. .,Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, #05-COR, Singapore, 117456, Singapore. .,National University of Singapore (Suzhou) Research Institute (NUSRI), Wuzhong Dist., Suzhou, Jiangsu Province, China.
| |
Collapse
|
4
|
de Lázaro I, Yilmazer A, Nam Y, Qubisi S, Razak FMA, Degens H, Cossu G, Kostarelos K. Non-viral, Tumor-free Induction of Transient Cell Reprogramming in Mouse Skeletal Muscle to Enhance Tissue Regeneration. Mol Ther 2018; 27:59-75. [PMID: 30470628 DOI: 10.1016/j.ymthe.2018.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 01/14/2023] Open
Abstract
Overexpression of Oct3/4, Klf4, Sox2, and c-Myc (OKSM) transcription factors can de-differentiate adult cells in vivo. While sustained OKSM expression triggers tumorigenesis through uncontrolled proliferation of toti- and pluripotent cells, transient reprogramming induces pluripotency-like features and proliferation only temporarily, without teratomas. We sought to transiently reprogram cells within mouse skeletal muscle with a localized injection of plasmid DNA encoding OKSM (pOKSM), and we hypothesized that the generation of proliferative intermediates would enhance tissue regeneration after injury. Intramuscular pOKSM administration rapidly upregulated pluripotency (Nanog, Ecat1, and Rex1) and early myogenesis genes (Pax3) in the healthy gastrocnemius of various strains. Mononucleated cells expressing such markers appeared in clusters among myofibers, proliferated only transiently, and did not lead to dysplasia or tumorigenesis for at least 120 days. Nanog was also upregulated in the gastrocnemius when pOKSM was administered 7 days after surgically sectioning its medial head. Enhanced tissue regeneration after reprogramming was manifested by the accelerated appearance of centronucleated myofibers and reduced fibrosis. These results suggest that transient in vivo reprogramming could develop into a novel strategy toward the acceleration of tissue regeneration after injury, based on the induction of transiently proliferative, pluripotent-like cells in situ. Further research to achieve clinically meaningful functional regeneration is warranted.
Collapse
Affiliation(s)
- Irene de Lázaro
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK; UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), London WC1N 1AX, UK
| | - Acelya Yilmazer
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK
| | - Yein Nam
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK; UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), London WC1N 1AX, UK
| | - Sara Qubisi
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK; UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), London WC1N 1AX, UK
| | - Fazilah Maizatul Abdul Razak
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK; UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), London WC1N 1AX, UK
| | - Hans Degens
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Michael Smith Building, The University of Manchester, Manchester M13 9PL, UK
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, UK; UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), London WC1N 1AX, UK.
| |
Collapse
|
5
|
Schukur L, Fussenegger M. Engineering of synthetic gene circuits for (re-)balancing physiological processes in chronic diseases. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:402-22. [DOI: 10.1002/wsbm.1345] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Lina Schukur
- Department of Biosystems Science and Engineering; ETH Zurich; Basel Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering; ETH Zurich; Basel Switzerland
- Faculty of Science; University of Basel; Basel Switzerland
| |
Collapse
|
6
|
Münst B, Thier MC, Winnemöller D, Helfen M, Thummer RP, Edenhofer F. Nanog induces suppression of senescence through downregulation of p27KIP1 expression. J Cell Sci 2016; 129:912-20. [PMID: 26795560 PMCID: PMC4813312 DOI: 10.1242/jcs.167932] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/04/2016] [Indexed: 02/05/2023] Open
Abstract
A comprehensive analysis of the molecular network of cellular factors establishing and maintaining pluripotency as well as self renewal of pluripotent stem cells is key for further progress in understanding basic stem cell biology. Nanog is necessary for the natural induction of pluripotency in early mammalian development but dispensable for both its maintenance and its artificial induction. To gain further insight into the molecular activity of Nanog, we analyzed the outcomes of Nanog gain-of-function in various cell models employing a recently developed biologically active recombinant cell-permeant protein, Nanog-TAT. We found that Nanog enhances the proliferation of both NIH 3T3 and primary fibroblast cells. Nanog transduction into primary fibroblasts results in suppression of senescence-associated β-galactosidase activity. Investigation of cell cycle factors revealed that transient activation of Nanog correlates with consistent downregulation of the cell cycle inhibitor p27KIP1 (also known as CDKN1B). By performing chromatin immunoprecipitation analysis, we confirmed bona fide Nanog-binding sites upstream of the p27KIP1 gene, establishing a direct link between physical occupancy and functional regulation. Our data demonstrates that Nanog enhances proliferation of fibroblasts through transcriptional regulation of cell cycle inhibitor p27 gene. Summary: Nanog blocks cellular senescence of fibroblasts through transcriptional regulation of cell cycle inhibitor p27KIP1.
Collapse
Affiliation(s)
- Bernhard Münst
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany
| | - Marc Christian Thier
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany
| | - Dirk Winnemöller
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany
| | - Martina Helfen
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany
| | - Rajkumar P Thummer
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Frank Edenhofer
- Stem Cell Engineering Group, Institute of Reconstructive Neurobiology, University of Bonn - Life & Brain Center and Hertie Foundation, Sigmund-Freud Str. 25, Bonn 53127, Germany Stem Cell and Regenerative Medicine Group, Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, Würzburg 97070, Germany Department of Genomics, Stem Cell Biology & Regenerative Medicine, Institute of Molecular Biology, Leopold-Franzens-University Innsbruck, Technikerstraße 25, Innsbruck 6020, Austria
| |
Collapse
|
7
|
Kimbrel EA, Lanza R. Current status of pluripotent stem cells: moving the first therapies to the clinic. Nat Rev Drug Discov 2015; 14:681-92. [PMID: 26391880 DOI: 10.1038/nrd4738] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cells (PSCs) hold great promise for drug discovery and regenerative medicine owing to their ability to differentiate into any cell type in the body. After more than three decades of research, including delays due to the potential tumorigenicity of PSCs and inefficiencies in differentiation methods, the field is at a turning point, with a number of clinical trials across the globe now testing PSC-derived products in humans. Ocular diseases dominate these first-in-man trials, and Phase l/ll results are showing promising safety data as well as possible efficacy. In addition, the advent of induced PSC (iPSC) technology is enabling the development of a wide range of cell-based disease models from genetically predisposed patients, thereby facilitating drug discovery. In this Review, we discuss the recent progress and remaining challenges for the use of PSCs in regenerative medicine and drug development.
Collapse
Affiliation(s)
- Erin A Kimbrel
- Ocata Therapeutics, 33 Locke Drive, Marlborough, Massachusetts 01752, USA
| | - Robert Lanza
- Ocata Therapeutics, 33 Locke Drive, Marlborough, Massachusetts 01752, USA
| |
Collapse
|
8
|
Abstract
Traditionally, cell analysis has focused on using molecular biomarkers for basic research, cell preparation, and clinical diagnostics; however, new microtechnologies are enabling evaluation of the mechanical properties of cells at throughputs that make them amenable to widespread use. We review the current understanding of how the mechanical characteristics of cells relate to underlying molecular and architectural changes, describe how these changes evolve with cell-state and disease processes, and propose promising biomedical applications that will be facilitated by the increased throughput of mechanical testing: from diagnosing cancer and monitoring immune states to preparing cells for regenerative medicine. We provide background about techniques that laid the groundwork for the quantitative understanding of cell mechanics and discuss current efforts to develop robust techniques for rapid analysis that aim to implement mechanophenotyping as a routine tool in biomedicine. Looking forward, we describe additional milestones that will facilitate broad adoption, as well as new directions not only in mechanically assessing cells but also in perturbing them to passively engineer cell state.
Collapse
Affiliation(s)
- Eric M Darling
- Center for Biomedical Engineering.,Department of Molecular Pharmacology, Physiology, and Biotechnology.,Department of Orthopaedics, and.,School of Engineering, Brown University, Providence, Rhode Island 02912;
| | - Dino Di Carlo
- Department of Bioengineering.,California NanoSystems Institute, and.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095;
| |
Collapse
|
9
|
Chen C, Dubin R, Kim MC. Emerging trends and new developments in regenerative medicine: a scientometric update (2000 - 2014). Expert Opin Biol Ther 2014; 14:1295-317. [PMID: 25077605 DOI: 10.1517/14712598.2014.920813] [Citation(s) in RCA: 360] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Our previous scientometric review of regenerative medicine provides a snapshot of the fast-growing field up to the end of 2011. The new review identifies emerging trends and new developments appearing in the literature of regenerative medicine based on relevant articles and reviews published between 2000 and the first month of 2014. AREAS COVERED Multiple datasets of publications relevant to regenerative medicine are constructed through topic search and citation expansion to ensure adequate coverage of the field. Networks of co-cited references representing the literature of regenerative medicine are constructed and visualized based on a combined dataset of 71,393 articles published between 2000 and 2014. Structural and temporal dynamics are identified in terms of most active topical areas and cited references. New developments are identified in terms of newly emerged clusters and research areas. Disciplinary-level patterns are visualized in dual-map overlays. EXPERT OPINION While research in induced pluripotent stem cells remains the most prominent area in the field of regenerative medicine, research related to clinical and therapeutic applications in regenerative medicine has experienced a considerable growth. In addition, clinical and therapeutic developments in regenerative medicine have demonstrated profound connections with the induced pluripotent stem cell research and stem cell research in general. A rapid adaptation of graphene-based nanomaterials in regenerative medicine is evident. Both basic research represented by stem cell research and application-oriented research typically found in tissue engineering are now increasingly integrated in the scientometric landscape of regenerative medicine. Tissue engineering is an interdisciplinary field in its own right. Advances in multiple disciplines such as stem cell research and graphene research have strengthened the connections between tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Chaomei Chen
- Drexel University, College of Computing and Informatics , 3141 Chestnut Street, Philadelphia, PA 19104-2875 , USA
| | | | | |
Collapse
|
10
|
Koh S, Piedrahita JA. From "ES-like" cells to induced pluripotent stem cells: a historical perspective in domestic animals. Theriogenology 2014; 81:103-11. [PMID: 24274415 DOI: 10.1016/j.theriogenology.2013.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 01/10/2023]
Abstract
Pluripotent stem cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) provide great potential as cell sources for gene editing to generate genetically modified animals, as well as in the field of regenerative medicine. Stable, long-term ESCs have been established in laboratory mouse and rat; however, isolation of true pluripotent ESCs in domesticated animals such as pigs and dogs have been less successful. Initially, domesticated animal pluripotent cell lines were referred to as "embryonic stem-like" cells owing to their similar morphologic characteristics to mouse ESCs, but accompanied by a limited ability to proliferate in vitro in an undifferentiated state. That is, they shared some but not all the characteristics of true ESCs. More recently, advances in reprogramming using exogenous transcription factors, combined with the utilization of small chemical inhibitors of key biochemical pathways, have led to the isolation of iPSCs. In this review, we provide a historical perspective of the isolation of various types of pluripotent stem cells in domesticated animals. In addition, we summarize the latest progress and limitations in the derivation and application of iPSCs.
Collapse
Affiliation(s)
- Sehwon Koh
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, USA; Genomics Program, North Carolina State University, Raleigh, North Carolina, USA
| | | |
Collapse
|
11
|
Gunewardene N, Bergen NV, Crombie D, Needham K, Dottori M, Nayagam BA. Directing human induced pluripotent stem cells into a neurosensory lineage for auditory neuron replacement. Biores Open Access 2014; 3:162-75. [PMID: 25126480 PMCID: PMC4120935 DOI: 10.1089/biores.2014.0019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Emerging therapies for sensorineural hearing loss include replacing damaged auditory neurons (ANs) using stem cells. Ultimately, it is important that these replacement cells can be patient-matched to avoid immunorejection. As human induced pluripotent stem cells (hiPSCs) can be obtained directly from the patient, they offer an opportunity to generate patient-matched neurons for transplantation. Here, we used an established neural induction protocol to differentiate two hiPSC lines (iPS1 and iPS2) and one human embryonic stem cell line (hESC; H9) toward a neurosensory lineage in vitro. Immunocytochemistry and qRT-PCR were used to analyze the expression of key markers involved in AN development at defined time points of differentiation. The hiPSC- and hESC-derived neurosensory progenitors expressed the dorsal hindbrain marker (PAX7), otic placodal marker (PAX2), proneurosensory marker (SOX2), ganglion neuronal markers (NEUROD1, BRN3A, ISLET1, ßIII-tubulin, Neurofilament kDa 160), and sensory AN markers (GATA3 and VGLUT1) over the time course examined. The hiPSC- and hESC-derived neurosensory progenitors had the highest expression levels of the sensory neural markers at 35 days in vitro. Furthermore, the neurons generated from this assay were found to be electrically active. While all cell lines analyzed produced functional neurosensory-like progenitors, variabilities in the levels of marker expression were observed between hiPSC lines and within samples of the same cell line, when compared with the hESC controls. Overall, these findings indicate that this neural assay was capable of differentiating hiPSCs toward a neurosensory lineage but emphasize the need for improving the consistency in the differentiation of hiPSCs into the required lineages.
Collapse
Affiliation(s)
- Niliksha Gunewardene
- Department of Otolaryngology, University of Melbourne , East Melbourne, Victoria, Australia
| | - Nicole Van Bergen
- Centre for Eye Research Australia, University of Melbourne , East Melbourne, Victoria, Australia
| | - Duncan Crombie
- Centre for Eye Research Australia, University of Melbourne , East Melbourne, Victoria, Australia
| | - Karina Needham
- Department of Otolaryngology, University of Melbourne , East Melbourne, Victoria, Australia
| | - Mirella Dottori
- Centre for Neural Engineering, University of Melbourne , Parkville, Victoria, Australia
| | - Bryony A Nayagam
- Centre for Eye Research Australia, University of Melbourne , East Melbourne, Victoria, Australia . ; Department of Audiology and Speech Pathology, University of Melbourne , Parkville, Victoria, Australia . ; Bionics Institute, University of Melbourne , East Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Induced pluripotent stem (iPS) cells: A new source for cell-based therapeutics? J Control Release 2014; 185:37-44. [DOI: 10.1016/j.jconrel.2014.04.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022]
|
13
|
In vivo cell reprogramming to pluripotency: exploring a novel tool for cell replenishment and tissue regeneration. Biochem Soc Trans 2014. [DOI: 10.1042/bst20140012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The potential of cell-replacement strategies for the treatment of disorders in which a particular cell type is damaged or degenerated has prompted the search for the perfect cell source. iPSCs (induced pluripotent stem cells) stand out as very advantageous candidates thanks to their self-renewal capacity and differentiation potential, together with the possibility of generating them from autologous somatic cells with minimally invasive techniques. However, their differentiation into the required cell type, precise delivery and successful engraftment and survival in the host are still challenging. We have proposed the transient reprogramming of somatic cells towards a pluripotent state in their in vivo microenvironment as a means to facilitate the regeneration of the tissue. The initial reports of in vivo reprogramming to pluripotency in the literature are reviewed and the potential clinical applications of this strategy are discussed.
Collapse
|
14
|
Human Pluripotent Stem Cell-Derived Retinal Pigmented Epithelium in Retinal Treatment: from Bench to Bedside. Mol Neurobiol 2014; 50:597-612. [DOI: 10.1007/s12035-014-8684-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/17/2014] [Indexed: 01/23/2023]
|
15
|
Koch PJ, Dinella J, Fete M, Siegfried EC, Koster MI. Modeling AEC-New approaches to study rare genetic disorders. Am J Med Genet A 2014; 164A:2443-54. [PMID: 24665072 DOI: 10.1002/ajmg.a.36455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/03/2014] [Indexed: 11/06/2022]
Abstract
Ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome is a rare monogenetic disorder that is characterized by severe abnormalities in ectoderm-derived tissues, such as skin and its appendages. A major cause of morbidity among affected infants is severe and chronic skin erosions. Currently, supportive care is the only available treatment option for AEC patients. Mutations in TP63, a gene that encodes key regulators of epidermal development, are the genetic cause of AEC. However, it is currently not clear how mutations in TP63 lead to the various defects seen in the patients' skin. In this review, we will discuss current knowledge of the AEC disease mechanism obtained by studying patient tissue and genetically engineered mouse models designed to mimic aspects of the disorder. We will then focus on new approaches to model AEC, including the use of patient cells and stem cell technology to replicate the disease in a human tissue culture model. The latter approach will advance our understanding of the disease and will allow for the development of new in vitro systems to identify drugs for the treatment of skin erosions in AEC patients. Further, the use of stem cell technology, in particular induced pluripotent stem cells (iPSC), will enable researchers to develop new therapeutic approaches to treat the disease using the patient's own cells (autologous keratinocyte transplantation) after correction of the disease-causing mutations.
Collapse
Affiliation(s)
- Peter J Koch
- Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado; Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado; Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, Colorado; Graduate Program in Cell Biology, Stem Cells and Development, University of Colorado School of Medicine, Aurora, Colorado
| | | | | | | | | |
Collapse
|
16
|
Mavroudi M, Zarogoulidis P, Porpodis K, Kioumis I, Lampaki S, Yarmus L, Malecki R, Zarogoulidis K, Malecki M. Stem cells' guided gene therapy of cancer: New frontier in personalized and targeted therapy. JOURNAL OF CANCER RESEARCH & THERAPY 2014; 2:22-33. [PMID: 24860662 PMCID: PMC4031908 DOI: 10.14312/2052-4994.2014-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Diagnosis and therapy of cancer remain to be the greatest challenges for all physicians working in clinical oncology and molecular medicine. The statistics speak for themselves with the grim reports of 1,638,910 men and women diagnosed with cancer and nearly 577,190 patients passed away due to cancer in the USA in 2012. For practicing clinicians, who treat patients suffering from advanced cancers with contemporary systemic therapies, the main challenge is to attain therapeutic efficacy, while minimizing side effects. Unfortunately, all contemporary systemic therapies cause side effects. In treated patients, these side effects may range from nausea to damaged tissues. In cancer survivors, the iatrogenic outcomes of systemic therapies may include genomic mutations and their consequences. Therefore, there is an urgent need for personalized and targeted therapies. Recently, we reviewed the current status of suicide gene therapy for cancer. Herein, we discuss the novel strategy: genetically engineered stem cells' guided gene therapy. REVIEW OF THERAPEUTIC STRATEGIES IN PRECLINICAL AND CLINICAL TRIALS Stem cells have the unique potential for self renewal and differentiation. This potential is the primary reason for introducing them into medicine to regenerate injured or degenerated organs, as well as to rejuvenate aging tissues. Recent advances in genetic engineering and stem cell research have created the foundations for genetic engineering of stem cells as the vectors for delivery of therapeutic transgenes. Specifically in oncology, the stem cells are genetically engineered to deliver the cell suicide inducing genes selectively to the cancer cells only. Expression of the transgenes kills the cancer cells, while leaving healthy cells unaffected. Herein, we present various strategies to bioengineer suicide inducing genes and stem cell vectors. Moreover, we review results of the main preclinical studies and clinical trials. However, the main risk for therapeutic use of stem cells is their cancerous transformation. Therefore, we discuss various strategies to safeguard stem cell guided gene therapy against iatrogenic cancerogenesis. PERSPECTIVES Defining cancer biomarkers to facilitate early diagnosis, elucidating cancer genomics and proteomics with modern tools of next generation sequencing, and analyzing patients' gene expression profiles provide essential data to elucidate molecular dynamics of cancer and to consider them for crafting pharmacogenomics-based personalized therapies. Streamlining of these data into genetic engineering of stem cells facilitates their use as the vectors delivering therapeutic genes into specific cancer cells. In this realm, stem cells guided gene therapy becomes a promising new frontier in personalized and targeted therapy of cancer.
Collapse
Affiliation(s)
- Maria Mavroudi
- “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU
| | - Paul Zarogoulidis
- “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU
| | - Konstantinos Porpodis
- “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU
| | - Ioannis Kioumis
- “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU
| | - Sofia Lampaki
- “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU
| | | | - Raf Malecki
- San Francisco State University, San Francisco, CA, USA
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA, USA
| | | | - Marek Malecki
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA, USA
- University of Wisconsin, Madison, WI, USA
| |
Collapse
|
17
|
Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv 2013; 31:1600-23. [PMID: 23962714 DOI: 10.1016/j.biotechadv.2013.08.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/20/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
Abstract
Recent technological advances in the generation, characterization, and bioprocessing of human pluripotent stem cells (hPSCs) have created new hope for their use as a source for production of cell-based therapeutic products. To date, a few clinical trials that have used therapeutic cells derived from hESCs have been approved by the Food and Drug Administration (FDA), but numerous new hPSC-based cell therapy products are under various stages of development in cell therapy-specialized companies and their future market is estimated to be very promising. However, the multitude of critical challenges regarding different aspects of hPSC-based therapeutic product manufacturing and their therapies have made progress for the introduction of new products and clinical applications very slow. These challenges include scientific, technological, clinical, policy, and financial aspects. The technological aspects of manufacturing hPSC-based therapeutic products for allogeneic and autologous cell therapies according to good manufacturing practice (cGMP) quality requirements is one of the most important challenging and emerging topics in the development of new hPSCs for clinical use. In this review, we describe main critical challenges and highlight a series of technological advances in all aspects of hPSC-based therapeutic product manufacturing including clinical grade cell line development, large-scale banking, upstream processing, downstream processing, and quality assessment of final cell therapeutic products that have brought hPSCs closer to clinical application and commercial cGMP manufacturing.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | |
Collapse
|
18
|
Pryzhkova MV. Concise review: carbon nanotechnology: perspectives in stem cell research. Stem Cells Transl Med 2013; 2:376-83. [PMID: 23572053 DOI: 10.5966/sctm.2012-0151] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Carbon nanotechnology has developed rapidly during the last decade, and carbon allotropes, especially graphene and carbon nanotubes, have already found a wide variety of applications in industry, high-tech fields, biomedicine, and basic science. Electroconductive nanomaterials have attracted great attention from tissue engineers in the design of remotely controlled cell-substrate interfaces. Carbon nanoconstructs are also under extensive investigation by clinical scientists as potential agents in anticancer therapies. Despite the recent progress in human pluripotent stem cell research, only a few attempts to use carbon nanotechnology in the stem cell field have been reported. However, acquired experience with and knowledge of carbon nanomaterials may be efficiently used in the development of future personalized medicine and in tissue engineering.
Collapse
|
19
|
Zarogoulidis P, Darwiche K, Sakkas A, Yarmus L, Huang H, Li Q, Freitag L, Zarogoulidis K, Malecki M. Suicide Gene Therapy for Cancer - Current Strategies. ACTA ACUST UNITED AC 2013; 4. [PMID: 24294541 DOI: 10.4172/2157-7412.1000139] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Current cancer treatments may create profound iatrogenic outcomes. The adverse effects of these treatments still remain, as the serious problems that practicing physicians have to cope with in clinical practice. Although, non-specific cytotoxic agents constitute an effective treatment modality against cancer cells, they also tend to kill normal, quickly dividing cells. On the other hand, therapies targeting the genome of the tumors are both under investigation, and some others are already streamlined to clinical practice. Several approaches have been investigated in order to find a treatment targeting the cancer cells, while not affecting the normal cells. Suicide gene therapy is a therapeutic strategy, in which cell suicide inducing transgenes are introduced into cancer cells. The two major suicide gene therapeutic strategies currently pursued are: cytosine deaminase/5-fluorocytosine and the herpes simplex virus/ganciclovir. The novel strategies include silencing gene expression, expression of intracellular antibodies blocking cells' vital pathways, and transgenic expression of caspases and DNases. We analyze various elements of cancer cells' suicide inducing strategies including: targets, vectors, and mechanisms. These strategies have been extensively investigated in various types of cancers, while exploring multiple delivery routes including viruses, non-viral vectors, liposomes, nanoparticles, and stem cells. We discuss various stages of streamlining of the suicide gene therapy into clinical oncology as applied to different types of cancer. Moreover, suicide gene therapy is in the center of attention as a strategy preventing cancer from developing in patients participating in the clinical trials of regenerative medicine. In oncology, these clinical trials are aimed at regenerating, with the aid of stem cells, of the patients' organs damaged by pathologic and/or iatrogenic factors. However, the stem cells carry the risk of neoplasmic transformation. We discuss cell suicide inducing strategies aimed at preventing stem cell-originated cancerogenesis.
Collapse
Affiliation(s)
- Paul Zarogoulidis
- Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece, EU ; Department of Interventional Pneumology, Ruhrlandklinik, West German Lung Center, University Hospital, University Duisburg-Essen, Essen, Germany, EU
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Malecki M, LaVanne C, Alhambra D, Dodivenaka C, Nagel S, Malecki R. Safeguarding Stem Cell-Based Regenerative Therapy against Iatrogenic Cancerogenesis: Transgenic Expression of DNASE1, DNASE1L3, DNASE2, DFFB Controlled By POLA1 Promoter in Proliferating and Directed Differentiation Resisting Human Autologous Pluripotent Induced Stem Cells Leads to their Death. ACTA ACUST UNITED AC 2013; Suppl 9. [PMID: 25045589 PMCID: PMC4103669 DOI: 10.4172/2157-7633.s9-005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction The worst possible complication of using stem cells for regenerative
therapy is iatrogenic cancerogenesis. The ultimate goal of our work is to
develop a self-triggering feedback mechanism aimed at causing death of all
stem cells, which resist directed differentiation, keep proliferating, and
can grow into tumors. Specific aim The specific aim was threefold: (1) to genetically engineer the DNA
constructs for the human, recombinant DNASE1, DNASE1L3, DNASE2,
DFFB controlled by POLA promoter; (2) to
bioengineer anti-SSEA-4 antibody guided vectors delivering transgenes to
human undifferentiated and proliferating pluripotent stem cells; (3) to
cause death of proliferating and directed differentiation resisting stem
cells by transgenic expression of the human recombinant the DNases
(hrDNases). Methods The DNA constructs for the human, recombinant DNASE1,
DNASE1L3, DNASE2, DFFB controlled by POLA
promoter were genetically engineered. The vectors targeting specifically
SSEA-4 expressing stem cells were bioengineered. The healthy
volunteers’ bone marrow mononuclear cells (BMMCs) were induced into
human, autologous, pluripotent stem cells with non-integrating plasmids.
Directed differentiation of the induced stem cells into endothelial cells
was accomplished with EGF and BMP. The anti-SSEA 4 antibodies’ guided
DNA vectors delivered the transgenes for the human recombinant
DNases’ into proliferating stem cells. Results Differentiation of the pluripotent induced stem cells into the
endothelial cells was verified by highlighting formation of tight and
adherens junctions through transgenic expression of recombinant fluorescent
fusion proteins: VE cadherin, claudin, zona occludens 1, and catenin.
Proliferation of the stem cells was determined through highlighting
transgenic expression of recombinant fluorescent proteins controlled by
POLA promoter, while also reporting expression of the
transgenes for the hrDNases. Expression of the transgenes for the DNases
resulted in complete collapse of the chromatin architecture and degradation
of the proliferating cells’ genomic DNA. The proliferating stem
cells, but not the differentiating ones, were effectively induced to
die. Conclusion Herein, we describe attaining the proof-of-concept for the strategy,
whereby transgenic expression of the genetically engineered human
recombinant DNases in proliferating and directed differentiation resisting
stem cells leads to their death. This novel strategy reduces the risk of
iatrogenic neoplasms in stem cell therapy.
Collapse
Affiliation(s)
- Marek Malecki
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA 94105, USA ; University of Wisconsin, Madison, WI 53706, USA
| | | | | | | | - Sarah Nagel
- South Dakota State University, Brookings, SD 57007, USA
| | - Raf Malecki
- Phoenix Biomolecular Engineering Foundation, San Francisco, CA 94105, USA ; San Francisco State University, San Francisco, CA 94123, USA
| |
Collapse
|
21
|
Adam Seger R, Actis P, Penfold C, Maalouf M, Vilozny B, Pourmand N. Voltage controlled nano-injection system for single-cell surgery. NANOSCALE 2012; 4:5843-6. [PMID: 22899383 PMCID: PMC4406976 DOI: 10.1039/c2nr31700a] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Manipulation and analysis of single cells is the next frontier in understanding processes that control the function and fate of cells. Herein we describe a single-cell injection platform based on nanopipettes. The system uses scanning microscopy techniques to detect cell surfaces, and voltage pulses to deliver molecules into individual cells. As a proof of concept, we injected adherent mammalian cells with fluorescent dyes.
Collapse
Affiliation(s)
- R Adam Seger
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Karumbayaram S, Lee P, Azghadi SF, Cooper AR, Patterson M, Kohn DB, Pyle A, Clark A, Byrne J, Zack JA, Plath K, Lowry WE. From skin biopsy to neurons through a pluripotent intermediate under Good Manufacturing Practice protocols. Stem Cells Transl Med 2011. [PMID: 23197638 DOI: 10.5966/sctm.2011-0001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The clinical application of human-induced pluripotent stem cells (hiPSCs) requires not only the production of Good Manufacturing Practice-grade (GMP-grade) hiPSCs but also the derivation of specified cell types for transplantation under GMP conditions. Previous reports have suggested that hiPSCs can be produced in the absence of animal-derived reagents (xenobiotics) to ease the transition to production under GMP standards. However, to facilitate the use of hiPSCs in cell-based therapeutics, their progeny should be produced not only in the absence of xenobiotics but also under GMP conditions requiring extensive standardization of protocols, documentation, and reproducibility of methods and product. Here, we present a successful framework to produce GMP-grade derivatives of hiPSCs that are free of xenobiotic exposure from the collection of patient fibroblasts, through reprogramming, maintenance of hiPSCs, identification of reprogramming vector integration sites (nrLAM-PCR), and finally specification and terminal differentiation of clinically relevant cells. Furthermore, we developed a primary set of Standard Operating Procedures for the GMP-grade derivation and differentiation of these cells as a resource to facilitate widespread adoption of these practices.
Collapse
|
24
|
Cheung C, Sinha S. Human embryonic stem cell-derived vascular smooth muscle cells in therapeutic neovascularisation. J Mol Cell Cardiol 2011; 51:651-64. [PMID: 21816157 DOI: 10.1016/j.yjmcc.2011.07.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 06/30/2011] [Accepted: 07/18/2011] [Indexed: 11/18/2022]
Abstract
Ischemic diseases remain one of the major causes of morbidity and mortality throughout the world. In recent clinical trials on cell-based therapies, the use of adult stem and progenitor cells only elicited marginal benefits. Therapeutic neovascularisation is the Holy Grail for ischemic tissue recovery. There is compelling evidence from animal transplantation studies that the inclusion of mural cells in addition to endothelial cells (ECs) can enhance the formation of functional blood vessels. Vascular smooth muscle cells (SMCs) and pericytes are essential for the stabilisation of nascent immature endothelial tubes. Despite the intense interest in the utility of human embryonic stem cells (ESCs) for vascular regenerative medicine, ESC-derived vascular SMCs have received much less attention than ECs. This review begins with developmental insights into a range of smooth muscle progenitors from studies on embryos and ESC differentiation systems. We then summarise the methods of derivation of smooth muscle progenitors and cells from human ESCs. The primary emphasis is on the inherent heterogeneity of smooth muscle progenitors and cells and the limitations of current in vitro characterisation. Essential transplantation issues such as the type and source of therapeutic cells, mode of cell delivery, measures to enhance cell viability, putative mechanisms of benefit and long-term tracking of cell fate are also discussed. Finally, we highlight the challenges of clinical compatibility and scaling up for medical use in order to eventually realise the goal of human ESC-based vascular regenerative medicine.
Collapse
Affiliation(s)
- Christine Cheung
- The Anne McLaren Laboratory for Regenerative Medicine, West Forvie Building, Forvie Site, University of Cambridge, Robinson Way, Cambridge CB2 0SZ, UK
| | | |
Collapse
|
25
|
Kiefer JC. Primer and interviews: Promises and realities of induced pluripotent stem cells. Dev Dyn 2011; 240:2034-41. [DOI: 10.1002/dvdy.22688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
26
|
Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, Mahairaki V, Koliatsos VE, Tung L, Zambidis ET. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS One 2011; 6:e18293. [PMID: 21494607 PMCID: PMC3072973 DOI: 10.1371/journal.pone.0018293] [Citation(s) in RCA: 309] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/23/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The production of cardiomyocytes from human induced pluripotent stem cells (hiPSC) holds great promise for patient-specific cardiotoxicity drug testing, disease modeling, and cardiac regeneration. However, existing protocols for the differentiation of hiPSC to the cardiac lineage are inefficient and highly variable. We describe a highly efficient system for differentiation of human embryonic stem cells (hESC) and hiPSC to the cardiac lineage. This system eliminated the variability in cardiac differentiation capacity of a variety of human pluripotent stem cells (hPSC), including hiPSC generated from CD34(+) cord blood using non-viral, non-integrating methods. METHODOLOGY/PRINCIPAL FINDINGS We systematically and rigorously optimized >45 experimental variables to develop a universal cardiac differentiation system that produced contracting human embryoid bodies (hEB) with an improved efficiency of 94.7±2.4% in an accelerated nine days from four hESC and seven hiPSC lines tested, including hiPSC derived from neonatal CD34(+) cord blood and adult fibroblasts using non-integrating episomal plasmids. This cost-effective differentiation method employed forced aggregation hEB formation in a chemically defined medium, along with staged exposure to physiological (5%) oxygen, and optimized concentrations of mesodermal morphogens BMP4 and FGF2, polyvinyl alcohol, serum, and insulin. The contracting hEB derived using these methods were composed of high percentages (64-89%) of cardiac troponin I(+) cells that displayed ultrastructural properties of functional cardiomyocytes and uniform electrophysiological profiles responsive to cardioactive drugs. CONCLUSION/SIGNIFICANCE This efficient and cost-effective universal system for cardiac differentiation of hiPSC allows a potentially unlimited production of functional cardiomyocytes suitable for application to hPSC-based drug development, cardiac disease modeling, and the future generation of clinically-safe nonviral human cardiac cells for regenerative medicine.
Collapse
Affiliation(s)
- Paul W. Burridge
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| | - Susan Thompson
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Michal A. Millrod
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Seth Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Xuan Yuan
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Ann Peters
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vasiliki Mahairaki
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vassilis E. Koliatsos
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- Department of Neurology, The Johns Hopkins University School of Medicine,
Baltimore, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Elias T. Zambidis
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| |
Collapse
|
27
|
Belay E, Mátrai J, Acosta-Sanchez A, Ma L, Quattrocelli M, Mátés L, Sancho-Bru P, Geraerts M, Yan B, Vermeesch J, Rincón MY, Samara-Kuko E, Ivics Z, Verfaillie C, Sampaolesi M, Izsvák Z, Vandendriessche T, Chuah MKL. Novel hyperactive transposons for genetic modification of induced pluripotent and adult stem cells: a nonviral paradigm for coaxed differentiation. Stem Cells 2011; 28:1760-71. [PMID: 20715185 DOI: 10.1002/stem.501] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adult stem cells and induced pluripotent stem cells (iPS) hold great promise for regenerative medicine. The development of robust nonviral approaches for stem cell gene transfer would facilitate functional studies and potential clinical applications. We have previously generated hyperactive transposases derived from Sleeping Beauty, using an in vitro molecular evolution and selection paradigm. We now demonstrate that these hyperactive transposases resulted in superior gene transfer efficiencies and expression in mesenchymal and muscle stem/progenitor cells, consistent with higher expression levels of therapeutically relevant proteins including coagulation factor IX. Their differentiation potential and karyotype was not affected. Moreover, stable transposition could also be achieved in iPS, which retained their ability to differentiate along neuronal, cardiac, and hepatic lineages without causing cytogenetic abnormalities. Most importantly, transposon-mediated delivery of the myogenic PAX3 transcription factor into iPS coaxed their differentiation into MYOD(+) myogenic progenitors and multinucleated myofibers, suggesting that PAX3 may serve as a myogenic "molecular switch" in iPS. Hence, this hyperactive transposon system represents an attractive nonviral gene transfer platform with broad implications for regenerative medicine, cell and gene therapy.
Collapse
Affiliation(s)
- Eyayu Belay
- Flanders Institute for Biotechnology (VIB), Vesalius Research Center, University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Asgari S, Pournasr B, Salekdeh GH, Ghodsizadeh A, Ott M, Baharvand H. Induced pluripotent stem cells: a new era for hepatology. J Hepatol 2010; 53:738-51. [PMID: 20621379 DOI: 10.1016/j.jhep.2010.05.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 05/09/2010] [Accepted: 05/13/2010] [Indexed: 12/17/2022]
Abstract
Stem cell transplantation has been proposed as an attractive alternative approach to restore liver mass and function. Recent progress has been reported on the generation of induced pluripotent stem (iPS) cells from somatic cells. Human-iPS cells can be differentiated towards the hepatic lineage which presents possibilities for improving research on diseases, drug development, tissue engineering, the development of bio-artificial livers, and a foundation for producing autologous cell therapies that would avoid immune rejection and enable correction of gene defects prior to cell transplantation. This focused review will discuss how human iPS cell advances are likely to have an impact on hepatology.
Collapse
Affiliation(s)
- Samira Asgari
- Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | | | | | | | | |
Collapse
|