151
|
Pellegrini S, Manenti F, Chimienti R, Nano R, Ottoboni L, Ruffini F, Martino G, Ravassard P, Piemonti L, Sordi V. Differentiation of Sendai Virus-Reprogrammed iPSC into β Cells, Compared with Human Pancreatic Islets and Immortalized β Cell Line. Cell Transplant 2018; 27:1548-1560. [PMID: 30251567 PMCID: PMC6180725 DOI: 10.1177/0963689718798564] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: New sources of insulin-secreting cells are strongly in demand for treatment
of diabetes. Induced pluripotent stem cells (iPSCs) have the potential to
generate insulin-producing cells (iβ). However, the gene expression profile
and secretory function of iβ still need to be validated in comparison with
native β cells. Methods: Two clones of human iPSCs, reprogrammed from adult fibroblasts through
integration-free Sendai virus, were differentiated into iβ and compared with
donor pancreatic islets and EndoC-βH1, an immortalized human β cell
line. Results: Both clones of iPSCs differentiated into insulin+ cells with high
efficiency (up to 20%). iβ were negative for pluripotency markers (Oct4,
Sox2, Ssea4) and positive for Pdx1, Nkx6.1, Chromogranin A, PC1/3, insulin,
glucagon and somatostatin. iβ basally secreted C-peptide, glucagon and
ghrelin and released insulin in response either to increasing concentration
of glucose or a depolarizing stimulus. The comparison revealed that iβ are
remarkably similar to donor derived islets in terms of gene and protein
expression profile and similar level of heterogeneity. The ability of iβ to
respond to glucose instead was more related to that of EndoC-βH1. Discussion: We demonstrated that insulin-producing cells generated from iPSCs
recapitulate fundamental gene expression profiles and secretory function of
native human β cells.
Collapse
Affiliation(s)
- Silvia Pellegrini
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Manenti
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raniero Chimienti
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rita Nano
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Linda Ottoboni
- 2 Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ruffini
- 2 Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- 2 Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,3 Vita-Salute San Raffaele University, Milan, Italy
| | - Philippe Ravassard
- 4 Institut du Cerveau et de la Moelle épinière (ICM), Biotechnology & Biotherapy Team, Université Pierre et Marie Curie, Paris, France
| | - Lorenzo Piemonti
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,3 Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Sordi
- 1 Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
152
|
Role of cell-secreted extracellular matrix formation in aggregate formation and stability of human induced pluripotent stem cells in suspension culture. J Biosci Bioeng 2018; 127:372-380. [PMID: 30249415 DOI: 10.1016/j.jbiosc.2018.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 11/22/2022]
Abstract
Clinical and industrial applications require large quantities of human induced pluripotent stem cells (hiPSCs); however, little is known regarding the mechanisms governing aggregate formation and stability in suspension culture. To address this, we determined differences in growth processes among hiPSC lines in suspension culture. Using an hiPSC aggregate suspension culture system, hiPSCs from different lines formed multicellular aggregates classified as large compact or small loose based on their size and morphology. Time-lapse observation of the growth processes of two different hiPSC lines revealed that the balance between cell division and the extent of subsequent cell death determined the final size and morphology of aggregates. Comparison of the cell survival and death of two hiPSC lines showed that the formation of small, loose aggregates was due to continued cell death during the exponential phase of growth, with apoptotic cells extruded from growing hiPSC aggregates by the concerted contraction of their neighbors. Western blot and immunofluorescent staining revealed that aggregate morphology and proliferative ability relied to a considerable extent upon secretion of the extracellular matrix (ECM). hiPSCs forming large compact and stable aggregates showed enhanced production of collagen type I in suspension culture at 120 h. Furthermore, these aggregates exhibited higher expression of E-cadherin and proliferation marker Ki-67 as compared with levels observed in small and loose aggregates at 120 h. These findings indicated that differences in both aggregate formation and stability in suspension culture among hiPSC lines were caused by differences in ECM secretion capacity.
Collapse
|
153
|
Ahmad R, Sportelli V, Ziller M, Spengler D, Hoffmann A. Tracing Early Neurodevelopment in Schizophrenia with Induced Pluripotent Stem Cells. Cells 2018; 7:E140. [PMID: 30227641 PMCID: PMC6162757 DOI: 10.3390/cells7090140] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/29/2022] Open
Abstract
Schizophrenia (SCZ) is a devastating mental disorder that is characterized by distortions in thinking, perception, emotion, language, sense of self, and behavior. Epidemiological evidence suggests that subtle perturbations in early neurodevelopment increase later susceptibility for disease, which typically manifests in adolescence to early adulthood. Early perturbations are thought to be significantly mediated through incompletely understood genetic risk factors. The advent of induced pluripotent stem cell (iPSC) technology allows for the in vitro analysis of disease-relevant neuronal cell types from the early stages of human brain development. Since iPSCs capture each donor's genotype, comparison between neuronal cells derived from healthy and diseased individuals can provide important insights into the molecular and cellular basis of SCZ. In this review, we discuss results from an increasing number of iPSC-based SCZ/control studies that highlight alterations in neuronal differentiation, maturation, and neurotransmission in addition to perturbed mitochondrial function and micro-RNA expression. In light of this remarkable progress, we consider also ongoing challenges from the field of iPSC-based disease modeling that call for further improvements on the generation and design of patient-specific iPSC studies to ultimately progress from basic studies on SCZ to tailored treatments.
Collapse
Affiliation(s)
- Ruhel Ahmad
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Vincenza Sportelli
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Michael Ziller
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Dietmar Spengler
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Anke Hoffmann
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
154
|
Southard SM, Kotipatruni RP, Rust WL. Generation and selection of pluripotent stem cells for robust differentiation to insulin-secreting cells capable of reversing diabetes in rodents. PLoS One 2018; 13:e0203126. [PMID: 30183752 PMCID: PMC6124757 DOI: 10.1371/journal.pone.0203126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 08/14/2018] [Indexed: 01/06/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology enables the creation and selection of pluripotent cells with specific genetic traits. This report describes a pluripotent cell line created specifically to form replacement pancreatic cells as a therapy for insulin-dependent diabetes. Beginning with primary pancreatic tissue acquired through organ donation, cells were isolated, re-programmed using non-integrating vectors and exposed to a four day differentiation protocol to generate definitive endoderm, a developmental precursor to pancreas. The best performing iPSC lines were then subjected to a 12-day basic differentiation protocol to generate endocrine pancreas precursors. The line that most consistently generated highly pure populations was selected for further development. This approach created an iPSC-variant cell line, SR1423, with a genetic profile correlated with preferential differentiation toward endodermal lineage at the loss of mesodermal potential. This report further describes an improved differentiation protocol that, coupled with SR1423, generated populations of greater than 60% insulin-expressing cells that secrete insulin in response to glucose and are capable of reversing diabetes in rodents. Created and banked following cGMP guidelines, SR1423 is a candidate cell line for the production of insulin-producing cells useful for the treatment of diabetes.
Collapse
|
155
|
Sonntag KC, Song B, Lee N, Jung JH, Cha Y, Leblanc P, Neff C, Kong SW, Carter BS, Schweitzer J, Kim KS. Pluripotent stem cell-based therapy for Parkinson's disease: Current status and future prospects. Prog Neurobiol 2018; 168:1-20. [PMID: 29653250 PMCID: PMC6077089 DOI: 10.1016/j.pneurobio.2018.04.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 03/13/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, which affects about 0.3% of the general population. As the population in the developed world ages, this creates an escalating burden on society both in economic terms and in quality of life for these patients and for the families that support them. Although currently available pharmacological or surgical treatments may significantly improve the quality of life of many patients with PD, these are symptomatic treatments that do not slow or stop the progressive course of the disease. Because motor impairments in PD largely result from loss of midbrain dopamine neurons in the substantia nigra pars compacta, PD has long been considered to be one of the most promising target diseases for cell-based therapy. Indeed, numerous clinical and preclinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells as a standardized therapeutic regimen has been fraught with fundamental ethical, practical, and clinical issues, prompting scientists to explore alternative cell sources. Based on groundbreaking establishments of human embryonic stem cells and induced pluripotent stem cells, these human pluripotent stem cells have been the subject of extensive research, leading to tremendous advancement in our understanding of these novel classes of stem cells and promising great potential for regenerative medicine. In this review, we discuss the prospects and challenges of human pluripotent stem cell-based cell therapy for PD.
Collapse
Affiliation(s)
- Kai-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Laboratory for Translational Research on Neurodegeneration, 115 Mill Street, Belmont, MA, 02478, United States; Program for Neuropsychiatric Research, 115 Mill Street, Belmont, MA, 02478, United States
| | - Bin Song
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Nayeon Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Jin Hyuk Jung
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Young Cha
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Pierre Leblanc
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Carolyn Neff
- Kaiser Permanente Medical Group, Irvine, CA, 92618, United States
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, United States; Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, United States
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States
| | - Jeffrey Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States.
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States.
| |
Collapse
|
156
|
O’Keefe CM, Pisanic TR, Zec H, Overman MJ, Herman JG, Wang TH. Facile profiling of molecular heterogeneity by microfluidic digital melt. SCIENCE ADVANCES 2018; 4:eaat6459. [PMID: 30263958 PMCID: PMC6157960 DOI: 10.1126/sciadv.aat6459] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/14/2018] [Indexed: 05/05/2023]
Abstract
This work presents a digital microfluidic platform called HYPER-Melt (high-density profiling and enumeration by melt) for highly parallelized copy-by-copy DNA molecular profiling. HYPER-Melt provides a facile means of detecting and assessing sequence variations of thousands of individual DNA molecules through digitization in a nanowell microchip array, allowing amplification and interrogation of individual template molecules by detecting HRM fluorescence changes due to sequence-dependent denaturation. As a model application, HYPER-Melt is used here for the detection and assessment of intermolecular heterogeneity of DNA methylation within the promoters of classical tumor suppressor genes. The capabilities of this platform are validated through serial dilutions of mixed epialleles, with demonstrated detection limits as low as 1 methylated variant in 2 million unmethylated templates (0.00005%) of a classic tumor suppressor gene, CDKN2A (p14ARF). The clinical potential of the platform is demonstrated using a digital assay for NDRG4, a tumor suppressor gene that is commonly methylated in colorectal cancer, in liquid biopsies of healthy and colorectal cancer patients. Overall, the platform provides the depth of information, simplicity of use, and single-molecule sensitivity necessary for rapid assessment of intermolecular variation contributing to genetic and epigenetic heterogeneity for challenging applications in embryogenesis, carcinogenesis, and rare biomarker detection.
Collapse
Affiliation(s)
- Christine M. O’Keefe
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD 21218, USA
| | - Helena Zec
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - James G. Herman
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Tza-Huei Wang
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD 21218, USA
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Corresponding author.
| |
Collapse
|
157
|
Yoon CH, Kim TW, Koh SJ, Choi YE, Hur J, Kwon YW, Cho HJ, Kim HS. Gata6 in pluripotent stem cells enhance the potential to differentiate into cardiomyocytes. BMB Rep 2018; 51:85-91. [PMID: 29335067 PMCID: PMC5836562 DOI: 10.5483/bmbrep.2018.51.2.176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Indexed: 01/15/2023] Open
Abstract
Pluripotent stem cell (PSC) variations can cause significant differences in the efficiency of cardiac differentiation. This process is unpredictable, as there is not an adequate indicator at the undifferentiated stage of the PSCs. We compared global gene expression profiles of two PSCs showing significant differences in cardiac differentiation potential. We identified 12 up-regulated genes related to heart development, and we found that 4 genes interacted with multiple genes. Among these genes, Gata6 is the only gene that was significantly induced at the early stage of differentiation of PSCs to cardiomyocytes. Gata6 knock-down in PSCs decreased the efficiency of cardiomyocyte production. In addition, we analyzed 6 mESC lines and 3 iPSC lines and confirmed that a positive correlation exists between Gata6 levels and efficiency of differentiation into cardiomyocytes. In conclusion, Gata6 could be utilized as a biomarker to select the best PSC lines to produce PSC-derived cardiomyocytes for therapeutic purposes. [BMB Reports 2018; 51(2): 85-91].
Collapse
Affiliation(s)
- Chang-Hwan Yoon
- Cardiovascular Center & Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Tae-Won Kim
- Molecular Medicine & Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
| | - Seok-Jin Koh
- Cardiovascular Center & Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Young-Eun Choi
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 03080, Korea
| | - Jin Hur
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 03080, Korea
| | - Yoo-Wook Kwon
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 03080, Korea
| | - Hyun-Jai Cho
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 03080, Korea
| | - Hyo-Soo Kim
- Molecular Medicine & Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
158
|
Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: Mechanisms of regulation and biotechnological applications. Biotechnol Adv 2018; 36:1946-1970. [PMID: 30077716 DOI: 10.1016/j.biotechadv.2018.08.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
The abilities of stem cells to self-renew and form different mature cells expand the possibilities of applications in cell-based therapies such as tissue recomposition in regenerative medicine, drug screening, and treatment of neurodegenerative diseases. In addition to stem cells found in the embryo, various adult organs and tissues have niches of stem cells in an undifferentiated state. In the central nervous system of adult mammals, neurogenesis occurs in two regions: the subventricular zone and the dentate gyrus in the hippocampus. The generation of the different neural lines originates in adult neural stem cells that can self-renew or differentiate into astrocytes, oligodendrocytes, or neurons in response to specific stimuli. The regulation of the fate of neural stem cells is a finely controlled process relying on a complex regulatory network that extends from the epigenetic to the translational level and involves extracellular matrix components. Thus, a better understanding of the mechanisms underlying how the process of neurogenesis is induced, regulated, and maintained will provide elues for development of novel for strategies for neurodegenerative therapies. In this review, we focus on describing the mechanisms underlying the regulation of the neuronal differentiation process by transcription factors, microRNAs, and extracellular matrix components.
Collapse
Affiliation(s)
- Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Anderson K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo C Parreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Alexandre H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil.
| |
Collapse
|
159
|
Abou-Saleh H, Zouein FA, El-Yazbi A, Sanoudou D, Raynaud C, Rao C, Pintus G, Dehaini H, Eid AH. The march of pluripotent stem cells in cardiovascular regenerative medicine. Stem Cell Res Ther 2018; 9:201. [PMID: 30053890 PMCID: PMC6062943 DOI: 10.1186/s13287-018-0947-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of global morbidity and mortality. Heart failure remains a major contributor to this mortality. Despite major therapeutic advances over the past decades, a better understanding of molecular and cellular mechanisms of CVD as well as improved therapeutic strategies for the management or treatment of heart failure are increasingly needed. Loss of myocardium is a major driver of heart failure. An attractive approach that appears to provide promising results in reducing cardiac degeneration is stem cell therapy (SCT). In this review, we describe different types of stem cells, including embryonic and adult stem cells, and we provide a detailed discussion of the properties of induced pluripotent stem cells (iPSCs). We also present and critically discuss the key methods used for converting somatic cells to pluripotent cells and iPSCs to cardiomyocytes (CMs), along with their advantages and limitations. Integrating and non-integrating reprogramming methods as well as characterization of iPSCs and iPSC-derived CMs are discussed. Furthermore, we critically present various methods of differentiating iPSCs to CMs. The value of iPSC-CMs in regenerative medicine as well as myocardial disease modeling and cardiac regeneration are emphasized.
Collapse
Affiliation(s)
- Haissam Abou-Saleh
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, “Attikon” Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christopher Rao
- Department of Surgery, Queen Elizabeth Hospital, Woolwich, London, UK
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Hassan Dehaini
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
160
|
Becherucci F, Mazzinghi B, Allinovi M, Angelotti ML, Romagnani P. Regenerating the kidney using human pluripotent stem cells and renal progenitors. Expert Opin Biol Ther 2018; 18:795-806. [PMID: 29939787 DOI: 10.1080/14712598.2018.1492546] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chronic kidney disease is a major health-care problem worldwide and its cost is becoming no longer affordable. Indeed, restoring damaged renal structures or building a new kidney represents an ambitious and ideal alternative to renal replacement therapy. Streams of research have explored the possible application of pluripotent stem cells (SCs) (embryonic SCs and induced pluripotent SCs) in different strategies aimed at regenerate functioning nephrons and at understanding the mechanisms of kidney regeneration. AREAS COVERED In this review, we will focus on the main potential applications of human pluripotent SCs to kidney regeneration, including those leading to rebuilding new kidneys or part of them (organoids, scaffolds, biological microdevices) as well as those aimed at understanding the pathophysiological mechanisms of renal disease and regenerative processes (modeling of kidney disease, genome editing). Moreover, we will discuss the role of endogenous renal progenitors cells in order to understand and promote kidney regeneration, as an attractive alternative to pluripotent SCs. EXPERT OPINION Opportunities and pitfalls of all these strategies will be underlined, finally leading to the conclusion that a deeper knowledge of the biology of pluripotent SCs is mandatory, in order to allow us to hypothesize their clinical application.
Collapse
Affiliation(s)
- Francesca Becherucci
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Benedetta Mazzinghi
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Marco Allinovi
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Maria Lucia Angelotti
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Paola Romagnani
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy.,b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| |
Collapse
|
161
|
Masia F, Glen A, Stephens P, Langbein W, Borri P. Label-free quantitative chemical imaging and classification analysis of adipogenesis using mouse embryonic stem cells. JOURNAL OF BIOPHOTONICS 2018; 11:e201700219. [PMID: 29573183 DOI: 10.1002/jbio.201700219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/26/2018] [Indexed: 06/08/2023]
Abstract
Stem cells have received much attention recently for their potential utility in regenerative medicine. The identification of their differentiated progeny often requires complex staining procedures, and is challenging for intermediary stages which are a priori unknown. In this work, the ability of label-free quantitative coherent anti-Stokes Raman scattering (CARS) micro-spectroscopy to identify populations of intermediate cell states during the differentiation of murine embryonic stem cells into adipocytes is assessed. Cells were imaged at different days of differentiation by hyperspectral CARS, and images were analysed with an unsupervised factorization algorithm providing Raman-like spectra and spatially resolved maps of chemical components. Chemical decomposition combined with a statistical analysis of their spatial distributions provided a set of parameters that were used for classification analysis. The first 2 principal components of these parameters indicated 3 main groups, attributed to undifferentiated cells, cells differentiated into committed white pre-adipocytes, and differentiating cells exhibiting a distinct protein globular structure with adjacent lipid droplets. An unsupervised classification methodology was developed, separating undifferentiated cell from cells in other stages, using a novel method to estimate the optimal number of clusters. The proposed unsupervised classification pipeline of hyperspectral CARS data offers a promising new tool for automated cell sorting in lineage analysis.
Collapse
Affiliation(s)
- Francesco Masia
- School of Physics and Astronomy, Cardiff University, Cardiff, UK
| | - Adam Glen
- School of Dentistry, Cardiff University, Cardiff, UK
| | - Phil Stephens
- School of Dentistry, Cardiff University, Cardiff, UK
| | | | - Paola Borri
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
162
|
Yu L, Li J, Hong J, Takashima Y, Fujimoto N, Nakajima M, Yamamoto A, Dong X, Dang Y, Hou Y, Yang W, Minami I, Okita K, Tanaka M, Luo C, Tang F, Chen Y, Tang C, Kotera H, Liu L. Low Cell-Matrix Adhesion Reveals Two Subtypes of Human Pluripotent Stem Cells. Stem Cell Reports 2018; 11:142-156. [PMID: 30008324 PMCID: PMC6067523 DOI: 10.1016/j.stemcr.2018.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 01/06/2023] Open
Abstract
We show that a human pluripotent stem cell (hPSC) population cultured on a low-adhesion substrate developed two hPSC subtypes with different colony morphologies: flat and domed. Notably, the dome-like cells showed higher active proliferation capacity and increased several pluripotent genes’ expression compared with the flat monolayer cells. We further demonstrated that cell-matrix adhesion mediates the interaction between cell morphology and expression of KLF4 and KLF5 through a serum response factor (SRF)-based regulatory double loop. Our results provide a mechanistic view on the coupling among adhesion, stem cell morphology, and pluripotency, shedding light on the critical role of cell-matrix adhesion in the induction and maintenance of hPSC. Low-adhesion substrates reveal two different subtypes co-exist in the hPSC population SRF-based regulatory loop-coupled adhesion, cell morphology, and KLF4/5 expression The low-adhesion substrates are more suitable for high-pluripotency cell culture
Collapse
Affiliation(s)
- Leqian Yu
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Nanometrics Laboratory, Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Junjun Li
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Jiayin Hong
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yasuhiro Takashima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Nanae Fujimoto
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Nanometrics Laboratory, Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Minako Nakajima
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Nanometrics Laboratory, Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Akihisa Yamamoto
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Xiaofeng Dong
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yujiao Dang
- Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing 100876, China
| | - Yu Hou
- Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing 100876, China
| | - Wei Yang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Itsunari Minami
- Department of Cell Design for Tissue Construction Faculty of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Keisuke Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Motomu Tanaka
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Physical Chemistry of Biosystems, Institute for Physical Chemistry, Heidelberg University, Heidelberg D69120, Germany
| | - Chunxiong Luo
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing 100876, China
| | - Yong Chen
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Ecole Normale Supérieure, CNRS-ENS-UPMC UMR 8640, 24 Rue Lhomond, Paris 75005, France
| | - Chao Tang
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| | - Hidetoshi Kotera
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Nanometrics Laboratory, Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan.
| | - Li Liu
- Institutes for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan.
| |
Collapse
|
163
|
Viventi S, Dottori M. Modelling the dorsal root ganglia using human pluripotent stem cells: A platform to study peripheral neuropathies. Int J Biochem Cell Biol 2018; 100:61-68. [PMID: 29772357 DOI: 10.1016/j.biocel.2018.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/08/2018] [Accepted: 05/13/2018] [Indexed: 12/28/2022]
Abstract
Sensory neurons of the dorsal root ganglia (DRG) are the primary responders to stimuli inducing feelings of touch, pain, temperature, vibration, pressure and muscle tension. They consist of multiple subpopulations based on their morphology, molecular and functional properties. Our understanding of DRG sensory neurons has been predominantly driven by rodent studies and using transformed cell lines, whereas less is known about human sensory DRG neurons simply because of limited availability of human tissue. Although these previous studies have been fundamental for our understanding of the sensory system, it is imperative to profile human DRG subpopulations as it is becoming evident that human sensory neurons do not share the identical molecular and functional properties found in other species. Furthermore, there are wide range of diseases and disorders that directly/indirectly cause sensory neuronal degeneration or dysfunctionality. Having an in vitro source of human DRG sensory neurons is paramount for studying their development, unique neuronal properties and for accelerating regenerative therapies to treat sensory neuropathies. Here we review the major studies describing generation of DRG sensory neurons from human pluripotent stem cells and fibroblasts and the gaps that need to be addressed for using in vitro-generated human DRG neurons to model human DRG tissue.
Collapse
Affiliation(s)
- Serena Viventi
- Department of Biomedical Engineering, University of Melbourne, Australia
| | - Mirella Dottori
- Department of Biomedical Engineering, University of Melbourne, Australia; Illawarra Health and Medical Research Institute, Centre for Molecular and Medical Bioscience, University of Wollongong, Australia.
| |
Collapse
|
164
|
Tsugata T, Nikoh N, Kin T, Miyagi-Shiohira C, Nakashima Y, Saitoh I, Noguchi Y, Ueki H, Watanabe M, Kobayashi N, Shapiro AMJ, Noguchi H. Role of Egr1 on Pancreatic Endoderm Differentiation. CELL MEDICINE 2018; 10:2155179017733177. [PMID: 32634182 PMCID: PMC6172987 DOI: 10.1177/2155179017733177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/18/2017] [Accepted: 04/27/2017] [Indexed: 11/17/2022]
Abstract
The low efficiency of in vitro differentiation of human embryonic stem cells (hESCs) or human-induced pluripotent stem cells (iPSCs) into insulin-producing cells is a crucial hurdle for the clinical implementation of human pluripotent stem cells (PSCs). Our previous investigation into the key factors for the differentiation of PSCs into insulin-producing cells suggested that the expression of GATA binding protein 6 (GATA6) and Gremlin 1 (GREM1) and inhibition of early growth response protein 1 (Egr1) may be important factors. In this study, we investigated the role of Egr1 in pancreas development. The transfection of small interfering RNA (siRNA) of Egr1 in the early phase induced the differentiation of iPSCs derived from fibroblasts (FiPSCs) into pancreatic endoderm and insulin-producing cells. In contrast, the downregulation of Egr1 in the late phase suppressed the differentiation of FiPSCs into pancreatic endoderm and insulin-producing cells. In addition, the overexpression of Egr1 suppressed the differentiation of iPSCs derived from pancreatic cells into pancreatic endoderm and insulin-producing cells. These data suggest that the downregulation of Egr1 in the early phase can efficiently induce the differentiation of iPSCs into insulin-producing cells.
Collapse
Affiliation(s)
- Takako Tsugata
- Natural and Environmental Sciences Program, The Open University of Japan, Chiba, Japan
| | - Naruo Nikoh
- Natural and Environmental Sciences Program, The Open University of Japan, Chiba, Japan
| | - Tatsuya Kin
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Yasufumi Noguchi
- Department of Socio-environmental Design, Hiroshima International University, Hiroshima, Japan
| | - Hideo Ueki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
165
|
Omole AE, Fakoya AOJ. Ten years of progress and promise of induced pluripotent stem cells: historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ 2018; 6:e4370. [PMID: 29770269 PMCID: PMC5951134 DOI: 10.7717/peerj.4370] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka in 2006 was heralded as a major breakthrough of the decade in stem cell research. The ability to reprogram human somatic cells to a pluripotent embryonic stem cell-like state through the ectopic expression of a combination of embryonic transcription factors was greeted with great excitement by scientists and bioethicists. The reprogramming technology offers the opportunity to generate patient-specific stem cells for modeling human diseases, drug development and screening, and individualized regenerative cell therapy. However, fundamental questions have been raised regarding the molecular mechanism of iPSCs generation, a process still poorly understood by scientists. The efficiency of reprogramming of iPSCs remains low due to the effect of various barriers to reprogramming. There is also the risk of chromosomal instability and oncogenic transformation associated with the use of viral vectors, such as retrovirus and lentivirus, which deliver the reprogramming transcription factors by integration in the host cell genome. These challenges can hinder the therapeutic prospects and promise of iPSCs and their clinical applications. Consequently, extensive studies have been done to elucidate the molecular mechanism of reprogramming and novel strategies have been identified which help to improve the efficiency of reprogramming methods and overcome the safety concerns linked with iPSC generation. Distinct barriers and enhancers of reprogramming have been elucidated, and non-integrating reprogramming methods have been reported. Here, we summarize the progress and the recent advances that have been made over the last 10 years in the iPSC field, with emphasis on the molecular mechanism of reprogramming, strategies to improve the efficiency of reprogramming, characteristics and limitations of iPSCs, and the progress made in the applications of iPSCs in the field of disease modelling, drug discovery and regenerative medicine. Additionally, this study appraises the role of genomic editing technology in the generation of healthy iPSCs.
Collapse
Affiliation(s)
- Adekunle Ebenezer Omole
- Department of Basic Sciences, American University of Antigua College of Medicine, St. John's, Antigua
| | | |
Collapse
|
166
|
Kusumoto D, Lachmann M, Kunihiro T, Yuasa S, Kishino Y, Kimura M, Katsuki T, Itoh S, Seki T, Fukuda K. Automated Deep Learning-Based System to Identify Endothelial Cells Derived from Induced Pluripotent Stem Cells. Stem Cell Reports 2018; 10:1687-1695. [PMID: 29754958 PMCID: PMC5989816 DOI: 10.1016/j.stemcr.2018.04.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 01/12/2023] Open
Abstract
Deep learning technology is rapidly advancing and is now used to solve complex problems. Here, we used deep learning in convolutional neural networks to establish an automated method to identify endothelial cells derived from induced pluripotent stem cells (iPSCs), without the need for immunostaining or lineage tracing. Networks were trained to predict whether phase-contrast images contain endothelial cells based on morphology only. Predictions were validated by comparison to immunofluorescence staining for CD31, a marker of endothelial cells. Method parameters were then automatically and iteratively optimized to increase prediction accuracy. We found that prediction accuracy was correlated with network depth and pixel size of images to be analyzed. Finally, K-fold cross-validation confirmed that optimized convolutional neural networks can identify endothelial cells with high performance, based only on morphology. Neural networks were trained to spot endothelial cells on phase-contrast images Performance was correlated with network depth and pixel size of training images Optimized networks identify endothelial cells with high accuracy
Collapse
Affiliation(s)
- Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Mark Lachmann
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Kunihiro
- LE Development Department, R&D Division, Medical Business Group, Sony Imaging Products & Solutions Inc., 4-14-1 Asahi-cho, Atsugi-shi, Kanagawa 243-0014, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mai Kimura
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiomi Katsuki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shogo Itoh
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomohisa Seki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
167
|
Gupta SK, Wesolowska-Andersen A, Ringgaard AK, Jaiswal H, Song L, Hastoy B, Ingvorsen C, Taheri-Ghahfarokhi A, Magnusson B, Maresca M, Jensen RR, Beer NL, Fels JJ, Grunnet LG, Thomas MK, Gloyn AL, Hicks R, McCarthy MI, Hansson M, Honoré C. NKX6.1 induced pluripotent stem cell reporter lines for isolation and analysis of functionally relevant neuronal and pancreas populations. Stem Cell Res 2018; 29:220-231. [PMID: 29734117 DOI: 10.1016/j.scr.2018.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/10/2018] [Accepted: 04/21/2018] [Indexed: 12/30/2022] Open
Abstract
Recent studies have reported significant advances in the differentiation of human pluripotent stem cells to clinically relevant cell types such as the insulin producing beta-like cells and motor neurons. However, many of the current differentiation protocols lead to heterogeneous cell cultures containing cell types other than the targeted cell fate. Genetically modified human pluripotent stem cells reporting the expression of specific genes are of great value for differentiation protocol optimization and for the purification of relevant cell populations from heterogeneous cell cultures. Here we present the generation of human induced pluripotent stem cell (iPSC) lines with a GFP reporter inserted in the endogenous NKX6.1 locus. Characterization of the reporter lines demonstrated faithful GFP labelling of NKX6.1 expression during pancreas and motor neuron differentiation. Cell sorting and gene expression profiling by RNA sequencing revealed that NKX6.1-positive cells from pancreatic differentiations closely resemble human beta cells. Furthermore, functional characterization of the isolated cells demonstrated that glucose-stimulated insulin secretion is mainly confined to the NKX6.1-positive cells. We expect that the NKX6.1-GFP iPSC lines and the results presented here will contribute to the further refinement of differentiation protocols and characterization of hPSC-derived beta cells and motor neurons for disease modelling and cell replacement therapies.
Collapse
Affiliation(s)
- Shailesh Kumar Gupta
- Discovery Biology, Discovery Sciences IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| | | | - Anna K Ringgaard
- Department of Stem Cell Biology, Novo Nordisk A/S, DK-2760 Måløv, Denmark
| | - Himjyot Jaiswal
- Discovery Biology, Discovery Sciences IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Luyan Song
- Lilly Research Laboratories, 46285 Indianapolis, IN, USA
| | - Benoit Hastoy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | | | | | - Björn Magnusson
- Discovery Biology, Discovery Sciences IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marcello Maresca
- Discovery Biology, Discovery Sciences IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rikke R Jensen
- Department of Stem Cell Biology, Novo Nordisk A/S, DK-2760 Måløv, Denmark
| | - Nicola L Beer
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Johannes J Fels
- Research Bioanalysis, Novo Nordisk A/S, DK-2760 Måløv, Denmark
| | - Lars G Grunnet
- Department of Stem Cell Biology, Novo Nordisk A/S, DK-2760 Måløv, Denmark
| | | | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Ryan Hicks
- Discovery Biology, Discovery Sciences IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| | - Mattias Hansson
- Stem Cell Research, Novo Nordisk A/S, DK-2760 Måløv, Denmark
| | - Christian Honoré
- Department of Stem Cell Biology, Novo Nordisk A/S, DK-2760 Måløv, Denmark.
| |
Collapse
|
168
|
Development of new method to enrich human iPSC-derived renal progenitors using cell surface markers. Sci Rep 2018; 8:6375. [PMID: 29686294 PMCID: PMC5913312 DOI: 10.1038/s41598-018-24714-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/05/2018] [Indexed: 01/02/2023] Open
Abstract
Cell therapy using renal progenitors differentiated from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs) has the potential to significantly reduce the number of patients receiving dialysis therapy. However, the differentiation cultures may contain undifferentiated or undesired cell types that cause unwanted side effects, such as neoplastic formation, when transplanted into a body. Moreover, the hESCs/iPSCs are often genetically modified in order to isolate the derived renal progenitors, hampering clinical applications. To establish an isolation method for renal progenitors induced from hESCs/iPSCs without genetic modifications, we screened antibodies against cell surface markers. We identified the combination of four markers, CD9−CD140a+CD140b+CD271+, which could enrich OSR1+SIX2+ renal progenitors. Furthermore, these isolated cells ameliorated renal injury in an acute kidney injury (AKI) mouse model when used for cell therapy. These cells could contribute to the development of hiPSC-based cell therapy and disease modeling against kidney diseases.
Collapse
|
169
|
Abstract
Bipolar disease (BD) is one of the major public health burdens worldwide and more people are affected every year. Comprehensive genetic studies have associated thousands of single nucleotide polymorphisms (SNPs) with BD risk; yet, very little is known about their functional roles. Induced pluripotent stem cells (iPSCs) are powerful tools for investigating the relationship between genotype and phenotype in disease-relevant tissues and cell types. Neural cells generated from BD-specific iPSCs are thought to capture associated genetic risk factors, known and unknown, and to allow the analysis of their effects on cellular and molecular phenotypes. Interestingly, an increasing number of studies on BD-derived iPSCs report distinct alterations in neural patterning, postmitotic calcium signaling, and neuronal excitability. Importantly, these alterations are partly normalized by lithium, a first line treatment in BD. In light of these exciting findings, we discuss current challenges to the field of iPSC-based disease modelling and future steps to be taken in order to fully exploit the potential of this approach for the investigation of BD and the development of new therapies.
Collapse
|
170
|
Kim SI, Matsumoto T, Kagawa H, Nakamura M, Hirohata R, Ueno A, Ohishi M, Sakuma T, Soga T, Yamamoto T, Woltjen K. Microhomology-assisted scarless genome editing in human iPSCs. Nat Commun 2018; 9:939. [PMID: 29507284 PMCID: PMC5838097 DOI: 10.1038/s41467-018-03044-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/16/2018] [Indexed: 12/26/2022] Open
Abstract
Gene-edited induced pluripotent stem cells (iPSCs) provide relevant isogenic human disease models in patient-specific or healthy genetic backgrounds. Towards this end, gene targeting using antibiotic selection along with engineered point mutations remains a reliable method to enrich edited cells. Nevertheless, integrated selection markers obstruct scarless transgene-free gene editing. Here, we present a method for scarless selection marker excision using engineered microhomology-mediated end joining (MMEJ). By overlapping the homology arms of standard donor vectors, short tandem microhomologies are generated flanking the selection marker. Unique CRISPR-Cas9 protospacer sequences nested between the selection marker and engineered microhomologies are cleaved after gene targeting, engaging MMEJ and scarless excision. Moreover, when point mutations are positioned unilaterally within engineered microhomologies, both mutant and normal isogenic clones are derived simultaneously. The utility and fidelity of our method is demonstrated in human iPSCs by editing the X-linked HPRT1 locus and biallelic modification of the autosomal APRT locus, eliciting disease-relevant metabolic phenotypes.
Collapse
Affiliation(s)
- Shin-Il Kim
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Tomoko Matsumoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Harunobu Kagawa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Michiko Nakamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Ryoko Hirohata
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Ayano Ueno
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Maki Ohishi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.
- Hakubi Center for Advanced Research, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
171
|
The impact of growth factors on human induced pluripotent stem cells differentiation into cardiomyocytes. Life Sci 2018; 196:38-47. [DOI: 10.1016/j.lfs.2018.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 01/29/2023]
|
172
|
Terryn J, Tricot T, Gajjar M, Verfaillie C. Recent advances in lineage differentiation from stem cells: hurdles and opportunities? F1000Res 2018; 7:220. [PMID: 29552337 PMCID: PMC5829467 DOI: 10.12688/f1000research.12596.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2018] [Indexed: 12/14/2022] Open
Abstract
Pluripotent stem cells have the property of long-term self-renewal and the potential to give rise to descendants of the three germ layers and hence all mature cells in the human body. Therefore, they hold the promise of offering insight not only into human development but also for human disease modeling and regenerative medicine. However, the generation of mature differentiated cells that closely resemble their
in vivo counterparts remains challenging. Recent advances in single-cell transcriptomics and computational modeling of gene regulatory networks are revealing a better understanding of lineage commitment and are driving modern genome editing approaches. Additional modification of the chemical microenvironment, as well as the use of bioengineering tools to recreate the cellular, extracellular matrix, and physical characteristics of the niche wherein progenitors and mature cells reside, is now being used to further improve the maturation and functionality of stem cell progeny.
Collapse
Affiliation(s)
- Joke Terryn
- Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Belgium
| | - Tine Tricot
- Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Belgium
| | - Madhavsai Gajjar
- Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Belgium
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Belgium
| |
Collapse
|
173
|
Laminin 521 Stabilizes the Pluripotency Expression Pattern of Human Embryonic Stem Cells Initially Derived on Feeder Cells. Stem Cells Int 2018. [PMID: 29535778 PMCID: PMC5835285 DOI: 10.1155/2018/7127042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human embryonic stem (hES) cells represent an important tool to study early cell development. The previously described use of human recombinant laminin (LN) 521 represented a step forward in generating clinically safe culture conditions. To test the short-term effect of LN521 on cultured hES cells, five male hES cell lines were cultured on human foreskin fibroblasts (hFFs), Matrigel, LN521, and LN121 and characterized by qPCR, immunofluorescence analysis, as well as their potential for three-germ layer differentiation. Variations in gene expression related to pluripotency, stemness, and testicular cells at different passages and culture conditions were evaluated by qPCR. All cell lines expressed pluripotency markers at protein and RNA level and were able to differentiate into cell types of the three germ layers after being cultured on LN521 for nine passages. Reduction in variation of pluripotency marker expression could be observed after culturing the cells on LN521 for nine passages. hES cells cultured on LN521 exhibited less differentiation, faster cell growth, and attachment when compared to hES cells cultured on LN121 or Matrigel. Our results indicate a positive effect of LN521 in stabilizing pluripotency gene expression and might be the first step towards more controllable and robust culture conditions for hES cells.
Collapse
|
174
|
Transcriptome variations among human embryonic stem cell lines are associated with their differentiation propensity. PLoS One 2018; 13:e0192625. [PMID: 29444173 PMCID: PMC5812638 DOI: 10.1371/journal.pone.0192625] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/26/2018] [Indexed: 12/20/2022] Open
Abstract
Human embryonic stem cells (hESCs) have the potential to form any cell type in the body, making them attractive cell sources in drug screening, regenerative medicine, disease and developmental processes modeling. However, not all hESC lines have the equal potency to generate desired cell types in vitro. Significant variations have been observed for the differentiation efficiency of various human ESC lines. The precise underpinning molecular mechanisms are still unclear. In this work, we compared transcriptome variations of four hESC lines H7, HUES1, HUES8 and HUES9. We found that hESC lines have different gene expression profiles, and these differentially expressed genes (DEGs) are significantly enriched in developmental processes, such as ectodermal, mesodermal and endodermal development. The enrichment difference between hESC lines was consistent with its lineage bias. Among these DEGs, some pluripotency factors and genes involved in signaling transduction showed great variations as well. The pleiotropic functions of these genes in controlling hESC identity and early lineage specification, implicated that different hESC lines may utilize distinct balance mechanisms to maintain pluripotent state. When the balance is broken in a certain environment, gene expression variation between them could impact on their different lineage specification behavior.
Collapse
|
175
|
Single-cell RNA sequencing reveals metallothionein heterogeneity during hESC differentiation to definitive endoderm. Stem Cell Res 2018; 28:48-55. [PMID: 29427839 DOI: 10.1016/j.scr.2018.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 12/04/2017] [Accepted: 01/11/2018] [Indexed: 12/25/2022] Open
Abstract
Differentiation of human pluripotent stem cells towards definitive endoderm (DE) is the critical first step for generating cells comprising organs such as the gut, liver, pancreas and lung. This in-vitro differentiation process generates a heterogeneous population with a proportion of cells failing to differentiate properly and maintaining expression of pluripotency factors such as Oct4. RNA sequencing of single cells collected at four time points during a 4-day DE differentiation identified high expression of metallothionein genes in the residual Oct4-positive cells that failed to differentiate to DE. Using X-ray fluorescence microscopy and multi-isotope mass spectrometry, we discovered that high intracellular zinc level corresponds with persistent Oct4 expression and failure to differentiate. This study improves our understanding of the cellular heterogeneity during in-vitro directed differentiation and provides a valuable resource to improve DE differentiation efficiency.
Collapse
|
176
|
Keller A, Dziedzicka D, Zambelli F, Markouli C, Sermon K, Spits C, Geens M. Genetic and epigenetic factors which modulate differentiation propensity in human pluripotent stem cells. Hum Reprod Update 2018; 24:162-175. [PMID: 29377992 DOI: 10.1093/humupd/dmx042] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/23/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human pluripotent stem cell (hPSC) lines are known to have a bias in their differentiation. This gives individual cell lines a propensity to preferentially differentiate towards one germ layer or cell type over others. Chromosomal aberrations, mitochondrial mutations, genetic diversity and epigenetic variance are the main drivers of this phenomenon, and can lead to a wide range of phenotypes. OBJECTIVE AND RATIONALE Our aim is to provide a comprehensive overview of the different factors which influence differentiation propensity. Specifically, we sought to highlight known genetic variances and their mechanisms, in addition to more general observations from larger abnormalities. Furthermore, we wanted to provide an up-to-date list of a growing number of predictive indicators which are able to identify differentiation propensity before the initiation of differentiation. As differentiation propensity can lead to difficulties in both research as well as clinical translation, our thorough overview could be a useful tool. SEARCH METHODS Combinations of the following key words were applied as search criteria in the PubMed database: embryonic stem cells, induced pluripotent stem cells, differentiation propensity (also: potential, efficiency, capacity, bias, variability), epigenetics, chromosomal abnormalities, genetic aberrations, X chromosome inactivation, mitochondrial function, mitochondrial metabolism, genetic diversity, reprogramming, predictive marker, residual stem cell, clinic. Only studies in English were included, ranging from 2000 to 2017, with a majority ranging from 2010 to 1017. Further manuscripts were added from cross-references. OUTCOMES Differentiation propensity is affected by a wide variety of (epi)genetic factors. These factors clearly lead to a loss of differentiation capacity, preference towards certain cell types and oftentimes, phenotypes which begin to resemble cancer. Broad changes in (epi)genetics, such as aneuploidies or wide-ranging modifications to the epigenetic landscape tend to lead to extensive, less definite changes in differentiation capacity, whereas more specific abnormalities often have precise ramifications in which certain cell types become more preferential. Furthermore, there appears to be a greater, though often less considered, contribution to differentiation propensity by factors such as mitochondria and inherent genetic diversity. Varied differentiation capacity can also lead to potential consequences in the clinical translation of hPSC, including the occurrence of residual undifferentiated stem cells, and the transplantation of potentially transformed cells. WIDER IMPLICATIONS As hPSC continue to advance towards the clinic, our understanding of them progresses as well. As a result, the challenges faced become more numerous, but also more clear. If the transition to the clinic is to be achieved with a minimum number of potential setbacks, thorough evaluation of the cells will be an absolute necessity. Altered differentiation propensity represents at least one such hurdle, for which researchers and eventually clinicians will need to find solutions. Already, steps are being taken to tackle the issue, though further research will be required to evaluate any long-term risks it poses.
Collapse
Affiliation(s)
- Alexander Keller
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Dominika Dziedzicka
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Filippo Zambelli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Christina Markouli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Karen Sermon
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Claudia Spits
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Mieke Geens
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| |
Collapse
|
177
|
Abu-Bonsrah KD, Zhang D, Bjorksten AR, Dottori M, Newgreen DF. Generation of Adrenal Chromaffin-like Cells from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:134-150. [PMID: 29233551 PMCID: PMC5768882 DOI: 10.1016/j.stemcr.2017.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 11/29/2022] Open
Abstract
Adrenomedullary chromaffin cells are catecholamine (CA)-producing cells originating from trunk neural crest (NC) via sympathoadrenal progenitors (SAPs). We generated NC and SAPs from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) in vitro via BMP2/FGF2 exposure, ascertained by qPCR and immunoexpression of SOX10, ASCL1, TFAP2α, and PHOX2B, and by fluorescence-activated cell sorting selection for p75NTR and GD2, and confirmed their trunk-like HOX gene expression. We showed that continuing BMP4 and curtailing FGF2 in vitro, augmented with corticosteroid mimetic, induced these cells to upregulate the chromaffin cell-specific marker PNMT and other CA synthesis and storage markers, and we demonstrated noradrenaline and adrenaline by Faglu and high-performance liquid chromatography. We showed these human cells' SAP-like property of migration and differentiation into cells expressing chromaffin cell markers by implanting them into avian embryos in vivo and in chorio-allantoic membrane grafts. These cells have the potential for investigating differentiation of human chromaffin cells and for modeling diseases involving this cell type.
Collapse
Affiliation(s)
- Kwaku Dad Abu-Bonsrah
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia; Centre for Neural Engineering, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Dongcheng Zhang
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia
| | - Andrew R Bjorksten
- Department of Anaesthesia and Pain Management, The Royal Melbourne Hospital Grattan Street, Parkville, 3052 VIC, Australia
| | - Mirella Dottori
- Centre for Neural Engineering, University of Melbourne, Parkville, 3010 VIC, Australia; Department of Anatomy and Neurosciences, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Donald F Newgreen
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia.
| |
Collapse
|
178
|
Theunissen TW, Jaenisch R. Mechanisms of gene regulation in human embryos and pluripotent stem cells. Development 2018; 144:4496-4509. [PMID: 29254992 DOI: 10.1242/dev.157404] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pluripotent stem cells have broad utility in biomedical research and their molecular regulation has thus garnered substantial interest. While the principles that establish and regulate pluripotency have been well defined in the mouse, it has been difficult to extrapolate these insights to the human system due to species-specific differences and the distinct developmental identities of mouse versus human embryonic stem cells. In this Review, we examine genome-wide approaches to elucidate the regulatory principles of pluripotency in human embryos and stem cells, and highlight where differences exist in the regulation of pluripotency in mice and humans. We review recent insights into the nature of human pluripotent cells in vivo, obtained by the deep sequencing of pre-implantation embryos. We also present an integrated overview of the principal layers of global gene regulation in human pluripotent stem cells. Finally, we discuss the transcriptional and epigenomic remodeling events associated with cell fate transitions into and out of human pluripotency.
Collapse
Affiliation(s)
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
179
|
Besser RR, Ishahak M, Mayo V, Carbonero D, Claure I, Agarwal A. Engineered Microenvironments for Maturation of Stem Cell Derived Cardiac Myocytes. Am J Cancer Res 2018; 8:124-140. [PMID: 29290797 PMCID: PMC5743464 DOI: 10.7150/thno.19441] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Through the use of stem cell-derived cardiac myocytes, tissue-engineered human myocardial constructs are poised for modeling normal and diseased physiology of the heart, as well as discovery of novel drugs and therapeutic targets in a human relevant manner. This review highlights the recent bioengineering efforts to recapitulate microenvironmental cues to further the maturation state of newly differentiated cardiac myocytes. These techniques include long-term culture, co-culture, exposure to mechanical stimuli, 3D culture, cell-matrix interactions, and electrical stimulation. Each of these methods has produced various degrees of maturation; however, a standardized measure for cardiomyocyte maturation is not yet widely accepted by the scientific community.
Collapse
|
180
|
Hicks MR, Hiserodt J, Paras K, Fujiwara W, Eskin A, Jan M, Xi H, Young CS, Evseenko D, Nelson SF, Spencer MJ, Handel BV, Pyle AD. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs. Nat Cell Biol 2018; 20:46-57. [PMID: 29255171 PMCID: PMC5962356 DOI: 10.1038/s41556-017-0010-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/16/2017] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-β signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells.
Collapse
MESH Headings
- Adult
- Aged
- CRISPR-Cas Systems
- Cell Differentiation
- Dystrophin/genetics
- Dystrophin/metabolism
- Female
- Gene Editing
- Gene Expression Regulation, Developmental
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Male
- Middle Aged
- Muscle Development/genetics
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Myoblasts/cytology
- Myoblasts/metabolism
- Myosins/genetics
- Myosins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Michael R Hicks
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Julia Hiserodt
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Katrina Paras
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Wakana Fujiwara
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Ascia Eskin
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Majib Jan
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Courtney S Young
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Neurology, University of California, Los Angeles, CA, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA.
| |
Collapse
|
181
|
Loo LSW, Lau HH, Jasmen JB, Lim CS, Teo AKK. An arduous journey from human pluripotent stem cells to functional pancreatic β cells. Diabetes Obes Metab 2018; 20:3-13. [PMID: 28474496 DOI: 10.1111/dom.12996] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/29/2017] [Accepted: 05/01/2017] [Indexed: 12/11/2022]
Abstract
Type 1 and type 2 diabetes are caused by a destruction and decrease in the number of functional insulin-producing β cells, respectively; therefore, the generation of functional β cells from human embryonic stem cells and human induced pluripotent stem cells, collectively known as human pluripotent stem cells (hPSCs), for potential cell replacement therapy and disease modelling is an intensely investigated area. Recent scientific breakthroughs enabled derivation of large quantities of human pancreatic β-like cells in vitro, although with varied glucose-stimulated insulin secretion kinetics. In the present review, we comprehensively summarize, compare and critically analyze the intricacies of these developing technologies, including differentiation platforms, robustness of protocols, and methodologies used to characterize hPSC-derived β-like cells. We also discuss experimental issues that need to be resolved before these β-like cells can be used clinically.
Collapse
Affiliation(s)
- Larry Sai Weng Loo
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hwee Hui Lau
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Joanita Binte Jasmen
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Chang Siang Lim
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
182
|
Kotaka M, Toyoda T, Yasuda K, Kitano Y, Okada C, Ohta A, Watanabe A, Uesugi M, Osafune K. Adrenergic receptor agonists induce the differentiation of pluripotent stem cell-derived hepatoblasts into hepatocyte-like cells. Sci Rep 2017; 7:16734. [PMID: 29196668 PMCID: PMC5711806 DOI: 10.1038/s41598-017-16858-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 11/18/2017] [Indexed: 12/24/2022] Open
Abstract
Current induction methods of hepatocytes from human induced pluripotent stem cells (hiPSCs) are neither low cost nor stable. By screening a chemical library of 1,120 bioactive compounds and known drugs, we identified the α1-adrenergic receptor agonist methoxamine hydrochloride as a small molecule that promotes the differentiation of hiPSC-derived hepatoblasts into ALBUMIN+ hepatocyte-like cells. Other α1-adrenergic receptor agonists also induced the differentiation of hepatocyte-like cells, and an α1-receptor antagonist blocked the hepatic-inducing activity of methoxamine hydrochloride and that of the combination of hepatocyte growth factor (HGF) and Oncostatin M (OsM), two growth factors often used for the induction of hepatoblasts into hepatocyte-like cells. We also confirmed that treatment with methoxamine hydrochloride activates the signal transducer and activator of transcription 3 (STAT3) pathway downstream of IL-6 family cytokines including OsM. These findings allowed us to establish hepatic differentiation protocols for both mouse embryonic stem cells (mESCs) and hiPSCs using small molecules at the step from hepatoblasts into hepatocyte-like cells. The results of the present study suggest that α1-adrenergic agonists induce hepatocyte-like cells by working downstream of HGF and OsM to activate STAT3.
Collapse
Affiliation(s)
- Maki Kotaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Katsutaro Yasuda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuko Kitano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Chihiro Okada
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Mitsubishi Space Software Co., Ltd., 5-4-36 Tsukaguchi-honmachi, Amagasaki, Hyogo, 661-0001, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Motonari Uesugi
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Institute for Chemical Research, Kyoto University, Gokasho Uji-city, Kyoto, 611-0011, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
183
|
Daneshvar K, Pondick JV, Kim BM, Zhou C, York SR, Macklin JA, Abualteen A, Tan B, Sigova AA, Marcho C, Tremblay KD, Mager J, Choi MY, Mullen AC. DIGIT Is a Conserved Long Noncoding RNA that Regulates GSC Expression to Control Definitive Endoderm Differentiation of Embryonic Stem Cells. Cell Rep 2017; 17:353-365. [PMID: 27705785 DOI: 10.1016/j.celrep.2016.09.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 08/12/2016] [Accepted: 09/03/2016] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) exhibit diverse functions, including regulation of development. Here, we combine genome-wide mapping of SMAD3 occupancy with expression analysis to identify lncRNAs induced by activin signaling during endoderm differentiation of human embryonic stem cells (hESCs). We find that DIGIT is divergent to Goosecoid (GSC) and expressed during endoderm differentiation. Deletion of the SMAD3-occupied enhancer proximal to DIGIT inhibits DIGIT and GSC expression and definitive endoderm differentiation. Disruption of the gene encoding DIGIT and depletion of the DIGIT transcript reveal that DIGIT is required for definitive endoderm differentiation. In addition, we identify the mouse ortholog of DIGIT and show that it is expressed during development and promotes definitive endoderm differentiation of mouse ESCs. DIGIT regulates GSC in trans, and activation of endogenous GSC expression is sufficient to rescue definitive endoderm differentiation in DIGIT-deficient hESCs. Our study defines DIGIT as a conserved noncoding developmental regulator of definitive endoderm.
Collapse
Affiliation(s)
- Kaveh Daneshvar
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Joshua V Pondick
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Byeong-Moo Kim
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Chan Zhou
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Samuel R York
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jillian A Macklin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ameed Abualteen
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Bo Tan
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; School of Fundamental Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Alla A Sigova
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Chelsea Marcho
- Department of Veterinary and Animal Sciences, University of Massachusetts at Amherst, Amherst, MA 01003, USA
| | - Kimberly D Tremblay
- Department of Veterinary and Animal Sciences, University of Massachusetts at Amherst, Amherst, MA 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts at Amherst, Amherst, MA 01003, USA
| | - Michael Y Choi
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Alan C Mullen
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
184
|
Williams EO, Taylor AK, Bell EL, Lim R, Kim DM, Guarente L. Sirtuin 1 Promotes Deacetylation of Oct4 and Maintenance of Naive Pluripotency. Cell Rep 2017; 17:809-820. [PMID: 27732856 DOI: 10.1016/j.celrep.2016.09.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/14/2016] [Accepted: 09/15/2016] [Indexed: 01/07/2023] Open
Abstract
The enhancer landscape is dramatically restructured as naive preimplantation epiblasts transition to the post-implantation state of primed pluripotency. A key factor in this process is Otx2, which is upregulated during the early stages of this transition and ultimately recruits Oct4 to a different set of enhancers. In this study, we discover that the acetylation status of Oct4 regulates the induction of the primed pluripotency gene network. Maintenance of the naive state requires the NAD-dependent deacetylase, SirT1, which deacetylates Oct4. The activity of SirT1 is reduced during the naive-to-primed transition; Oct4 becomes hyper-acetylated and binds to an Otx2 enhancer to induce Otx2 expression. Induction of Otx2 causes the reorganization of acetylated Oct4 and results in the induction of the primed pluripotency gene network. Regulation of Oct4 by SirT1 may link stem cell development to environmental conditions, and it may provide strategies to manipulate epiblast cell state.
Collapse
Affiliation(s)
- Eric O Williams
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amy K Taylor
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric L Bell
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rachelle Lim
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel M Kim
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leonard Guarente
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
185
|
Naive-like ESRRB + iPSCs with the Capacity for Rapid Neural Differentiation. Stem Cell Reports 2017; 9:1825-1838. [PMID: 29129686 PMCID: PMC5785673 DOI: 10.1016/j.stemcr.2017.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022] Open
Abstract
Several groups have reported the existence of a form of pluripotency that resembles that of mouse embryonic stem cells (mESCs), i.e., a naive state, in human pluripotent stem cells; however, the characteristics vary between reports. The nuclear receptor ESRRB is expressed in mESCs and plays a significant role in their self-renewal, but its expression has not been observed in most naive-like human induced pluripotent stem cells (hiPSCs). In this study, we modified several methods for converting hiPSCs into a naive state through the transgenic expression of several reprogramming factors. The resulting cells express the components of the core transcriptional network of mESCs, including ESRRB, at high levels, which suggests the existence of naive-state hiPSCs that are similar to mESCs. We also demonstrate that these cells differentiate more readily into neural cells than do conventional hiPSCs. These features may be beneficial for their use in disease modeling and regenerative medicine.
Collapse
|
186
|
Fujimori K, Matsumoto T, Kisa F, Hattori N, Okano H, Akamatsu W. Escape from Pluripotency via Inhibition of TGF-β/BMP and Activation of Wnt Signaling Accelerates Differentiation and Aging in hPSC Progeny Cells. Stem Cell Reports 2017; 9:1675-1691. [PMID: 29107593 PMCID: PMC5831048 DOI: 10.1016/j.stemcr.2017.09.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a potentially valuable cell source for applications in cell replacement therapy, drug development, and disease modeling. For all these uses, it is necessary to develop reproducible and robust protocols for differentiation into desired cell types. However, differentiation protocols remain unstable and inefficient, which makes minimizing the differentiation variance among hPSC lines and obtaining purified terminally differentiated cells extremely time consuming. Here, we report a simple treatment with three small molecules—SB431542, dorsomorphine, and CHIR99021—that enhanced hPSC differentiation into three germ layers with a chemically transitional embryoid-body-like state (CTraS). Induction of CTraS reduced the innate differentiation propensities of hPSCs (even unfavorably differentiated hPSCs) and shifted their differentiation into terminally differentiated cells, particularly neurons. In addition, CTraS induction accelerated in vitro pathological expression concurrently with neural maturation. Thus, CTraS can promote the latent potential of hPSCs for differentiation and potentially expand the utility and applicability of hPSCs. CTraS induction enhances hPSC differentiation into three germ layers without bias CTraS induction is applicable to a wide range of hPSCs even without colony selection Developing a robust neural induction protocol via CTraS for hPSC disease modeling CTraS induction promotes in vitro pathological expression with maturation and aging
Collapse
Affiliation(s)
- Koki Fujimori
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; Research Fellow of Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Takuya Matsumoto
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Fumihiko Kisa
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Wado Akamatsu
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8431, Japan.
| |
Collapse
|
187
|
Xie AW, Binder BYK, Khalil AS, Schmitt SK, Johnson HJ, Zacharias NA, Murphy WL. Controlled Self-assembly of Stem Cell Aggregates Instructs Pluripotency and Lineage Bias. Sci Rep 2017; 7:14070. [PMID: 29070799 PMCID: PMC5656593 DOI: 10.1038/s41598-017-14325-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022] Open
Abstract
Stem cell-derived organoids and other 3D microtissues offer enormous potential as models for drug screening, disease modeling, and regenerative medicine. Formation of stem/progenitor cell aggregates is common in biomanufacturing processes and critical to many organoid approaches. However, reproducibility of current protocols is limited by reliance on poorly controlled processes (e.g., spontaneous aggregation). Little is known about the effects of aggregation parameters on cell behavior, which may have implications for the production of cell aggregates and organoids. Here we introduce a bioengineered platform of labile substrate arrays that enable simple, scalable generation of cell aggregates via a controllable 2D-to-3D "self-assembly". As a proof-of-concept, we show that labile substrates generate size- and shape-controlled embryoid bodies (EBs) and can be easily modified to control EB self-assembly kinetics. We show that aggregation method instructs EB lineage bias, with faster aggregation promoting pluripotency loss and ectoderm, and slower aggregation favoring mesoderm and endoderm. We also find that aggregation kinetics of EBs markedly influence EB structure, with slower kinetics resulting in increased EB porosity and growth factor signaling. Our findings suggest that controlling internal structure of cell aggregates by modifying aggregation kinetics is a potential strategy for improving 3D microtissue models for research and translational applications.
Collapse
Affiliation(s)
- Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Bernard Y K Binder
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Samantha K Schmitt
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Nicholas A Zacharias
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, United States.
| |
Collapse
|
188
|
Ortmann D, Vallier L. Variability of human pluripotent stem cell lines. Curr Opin Genet Dev 2017; 46:179-185. [DOI: 10.1016/j.gde.2017.07.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 12/23/2022]
|
189
|
Hitomi H, Kasahara T, Katagiri N, Hoshina A, Mae SI, Kotaka M, Toyohara T, Rahman A, Nakano D, Niwa A, Saito MK, Nakahata T, Nishiyama A, Osafune K. Human pluripotent stem cell–derived erythropoietin-producing cells ameliorate renal anemia in mice. Sci Transl Med 2017; 9:9/409/eaaj2300. [DOI: 10.1126/scitranslmed.aaj2300] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 04/27/2017] [Indexed: 11/02/2022]
|
190
|
Boroviak T, Nichols J. Primate embryogenesis predicts the hallmarks of human naïve pluripotency. Development 2017; 144:175-186. [PMID: 28096211 PMCID: PMC5430762 DOI: 10.1242/dev.145177] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Naïve pluripotent mouse embryonic stem cells (ESCs) resemble the preimplantation epiblast and efficiently contribute to chimaeras. Primate ESCs correspond to the postimplantation embryo and fail to resume development in chimaeric assays. Recent data suggest that human ESCs can be ‘reset’ to an earlier developmental stage, but their functional capacity remains ill defined. Here, we discuss how the naïve state is inherently linked to preimplantation epiblast identity in the embryo. We hypothesise that distinctive features of primate development provide stringent criteria to evaluate naïve pluripotency in human and other primate cells. Based on our hypothesis, we define 12 key hallmarks of naïve pluripotency, five of which are specific to primates. These hallmarks may serve as a functional framework to assess human naïve ESCs. Summary: This Hypothesis article highlights several fundamental differences between rodent and primate early development and exploits these to predict key hallmarks of naïve pluripotency in primates.
Collapse
Affiliation(s)
- Thorsten Boroviak
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.,Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 4BG, UK
| |
Collapse
|
191
|
IAP-Based Cell Sorting Results in Homogeneous Transplantable Dopaminergic Precursor Cells Derived from Human Pluripotent Stem Cells. Stem Cell Reports 2017; 9:1207-1220. [PMID: 28943253 PMCID: PMC5639383 DOI: 10.1016/j.stemcr.2017.08.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived mesencephalic dopaminergic (mesDA) neurons can relieve motor deficits in animal models of Parkinson's disease (PD). Clinical translation of differentiation protocols requires standardization of production procedures, and surface-marker-based cell sorting is considered instrumental for reproducible generation of defined cell products. Here, we demonstrate that integrin-associated protein (IAP) is a cell surface marker suitable for enrichment of hPSC-derived mesDA progenitor cells. Immunomagnetically sorted IAP+ mesDA progenitors showed increased expression of ventral midbrain floor plate markers, lacked expression of pluripotency markers, and differentiated into mature dopaminergic (DA) neurons in vitro. Intrastriatal transplantation of IAP+ cells sorted at day 16 of differentiation in a rat model of PD resulted in functional recovery. Grafts from sorted IAP+ mesDA progenitors were more homogeneous in size and DA neuron density. Thus, we suggest IAP-based sorting for reproducible prospective enrichment of mesDA progenitor cells in clinical cell replacement strategies. Flow cytometric antibody screening identified IAP as a marker for mesDA progenitors IAP+ cells displayed hallmark characteristics of ventral floor plate cells Immunomagnetic IAP sorting led to reproducible and homogeneous cell compositions IAP+ cells generated mature grafts leading to functional recovery in lesioned rats
Collapse
|
192
|
Peters DT, Henderson CA, Warren CR, Friesen M, Xia F, Becker CE, Musunuru K, Cowan CA. Asialoglycoprotein receptor 1 is a specific cell-surface marker for isolating hepatocytes derived from human pluripotent stem cells. Development 2017; 143:1475-81. [PMID: 27143754 DOI: 10.1242/dev.132209] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 02/29/2016] [Indexed: 12/14/2022]
Abstract
Hepatocyte-like cells (HLCs) are derived from human pluripotent stem cells (hPSCs) in vitro, but differentiation protocols commonly give rise to a heterogeneous mixture of cells. This variability confounds the evaluation of in vitro functional assays performed using HLCs. Increased differentiation efficiency and more accurate approximation of the in vivo hepatocyte gene expression profile would improve the utility of hPSCs. Towards this goal, we demonstrate the purification of a subpopulation of functional HLCs using the hepatocyte surface marker asialoglycoprotein receptor 1 (ASGR1). We analyzed the expression profile of ASGR1-positive cells by microarray, and tested their ability to perform mature hepatocyte functions (albumin and urea secretion, cytochrome activity). By these measures, ASGR1-positive HLCs are enriched for the gene expression profile and functional characteristics of primary hepatocytes compared with unsorted HLCs. We have demonstrated that ASGR1-positive sorting isolates a functional subpopulation of HLCs from among the heterogeneous cellular population produced by directed differentiation.
Collapse
Affiliation(s)
- Derek T Peters
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA Harvard Medical School, Boston, MA 02115, USA
| | - Christopher A Henderson
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Curtis R Warren
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Max Friesen
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Fang Xia
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Caroline E Becker
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Kiran Musunuru
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Chad A Cowan
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
193
|
Klein M, Onnink M, van Donkelaar M, Wolfers T, Harich B, Shi Y, Dammers J, Arias-Vásquez A, Hoogman M, Franke B. Brain imaging genetics in ADHD and beyond - Mapping pathways from gene to disorder at different levels of complexity. Neurosci Biobehav Rev 2017; 80:115-155. [PMID: 28159610 PMCID: PMC6947924 DOI: 10.1016/j.neubiorev.2017.01.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/08/2016] [Accepted: 01/09/2017] [Indexed: 01/03/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common and often persistent neurodevelopmental disorder. Beyond gene-finding, neurobiological parameters, such as brain structure, connectivity, and function, have been used to link genetic variation to ADHD symptomatology. We performed a systematic review of brain imaging genetics studies involving 62 ADHD candidate genes in childhood and adult ADHD cohorts. Fifty-one eligible research articles described studies of 13 ADHD candidate genes. Almost exclusively, single genetic variants were studied, mostly focussing on dopamine-related genes. While promising results have been reported, imaging genetics studies are thus far hampered by methodological differences in study design and analysis methodology, as well as limited sample sizes. Beyond reviewing imaging genetics studies, we also discuss the need for complementary approaches at multiple levels of biological complexity and emphasize the importance of combining and integrating findings across levels for a better understanding of biological pathways from gene to disease. These may include multi-modal imaging genetics studies, bioinformatic analyses, and functional analyses of cell and animal models.
Collapse
Affiliation(s)
- Marieke Klein
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Marten Onnink
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Marjolein van Donkelaar
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Thomas Wolfers
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Benjamin Harich
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Yan Shi
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Janneke Dammers
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Alejandro Arias-Vásquez
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Cognitive Neuroscience, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Martine Hoogman
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| |
Collapse
|
194
|
Pournasr B, Duncan SA. Modeling Inborn Errors of Hepatic Metabolism Using Induced Pluripotent Stem Cells. Arterioscler Thromb Vasc Biol 2017; 37:1994-1999. [PMID: 28818857 DOI: 10.1161/atvbaha.117.309199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/07/2017] [Indexed: 01/09/2023]
Abstract
Inborn errors of hepatic metabolism are because of deficiencies commonly within a single enzyme as a consequence of heritable mutations in the genome. Individually such diseases are rare, but collectively they are common. Advances in genome-wide association studies and DNA sequencing have helped researchers identify the underlying genetic basis of such diseases. Unfortunately, cellular and animal models that accurately recapitulate these inborn errors of hepatic metabolism in the laboratory have been lacking. Recently, investigators have exploited molecular techniques to generate induced pluripotent stem cells from patients' somatic cells. Induced pluripotent stem cells can differentiate into a wide variety of cell types, including hepatocytes, thereby offering an innovative approach to unravel the mechanisms underlying inborn errors of hepatic metabolism. Moreover, such cell models could potentially provide a platform for the discovery of therapeutics. In this mini-review, we present a brief overview of the state-of-the-art in using pluripotent stem cells for such studies.
Collapse
Affiliation(s)
- Behshad Pournasr
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston (B.P., S.A.D.); and Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran (B.P.)
| | - Stephen A Duncan
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston (B.P., S.A.D.); and Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran (B.P.).
| |
Collapse
|
195
|
Kondo Y, Toyoda T, Ito R, Funato M, Hosokawa Y, Matsui S, Sudo T, Nakamura M, Okada C, Zhuang X, Watanabe A, Ohta A, Inagaki N, Osafune K. Identification of a small molecule that facilitates the differentiation of human iPSCs/ESCs and mouse embryonic pancreatic explants into pancreatic endocrine cells. Diabetologia 2017; 60:1454-1466. [PMID: 28534195 DOI: 10.1007/s00125-017-4302-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/12/2017] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Pancreatic beta-like cells generated from human induced pluripotent stem cells (hiPSCs) or human embryonic stem cells (hESCs) offer an appealing donor tissue source. However, differentiation protocols that mainly use growth factors are costly. Therefore, in this study, we aimed to establish efficient differentiation protocols to change hiPSCs/hESCs to insulin (INS)+ cells using novel small-molecule inducers. METHODS We screened small molecules that increased the induction rate of INS+ cells from hESC-derived pancreatic and duodenal homeobox 1 (PDX1)+ pancreatic progenitor cells. The differentiation protocol to generate INS+ cells from hiPSCs/hESCs was optimised using hit compounds, and INS+ cells induced with the compounds were characterised for their in vitro and in vivo functions. The inducing activity of the hit compounds was also examined using mouse embryonic pancreatic tissues in an explant culture system. Finally, RNA sequencing analyses were performed on the INS+ cells to elucidate the mechanisms of action by which the hit compounds induced pancreatic endocrine differentiation. RESULTS One hit compound, sodium cromoglicate (SCG), was identified out of approximately 1250 small molecules screened. When SCG was combined with a previously described protocol, the induction rate of INS+ cells increased from a mean ± SD of 5.9 ± 1.5% (n = 3) to 16.5 ± 2.1% (n = 3). SCG induced neurogenin 3-positive cells at a mean ± SD of 32.6 ± 4.6% (n = 3) compared with 14.2 ± 3.6% (n = 3) for control treatment without SCG, resulting in an increased generation of endocrine cells including insulin-producing cells. Similar induction by SCG was confirmed using mouse embryonic pancreatic explants. We also confirmed that the mechanisms of action by which SCG induced pancreatic endocrine differentiation included the inhibition of bone morphogenetic protein 4 signalling. CONCLUSIONS/INTERPRETATION SCG improves the generation of pancreatic endocrine cells from multiple hiPSC/hESC lines and mouse embryonic pancreatic explants by facilitating the differentiation of endocrine precursors. This discovery will contribute to elucidating the mechanisms of pancreatic endocrine development and facilitate cost-effective generation of INS+ cells from hiPSCs/hESCs. DATA AVAILABILITY The RNA sequencing data generated during the current study are available in the Gene Expression Omnibus ( www.ncbi.nlm.nih.gov/geo ) with series accession number GSE89973.
Collapse
Affiliation(s)
- Yasushi Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Michinori Funato
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshiya Hosokawa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Satoshi Matsui
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomomi Sudo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Nakamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Chihiro Okada
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Mitsubishi Space Software Co., Ltd, 5-4-36, Tsukaguchi-honmachi, Amagasaki, Hyogo, Japan
| | - Xiaotong Zhuang
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
196
|
Son B, Kim JA, Cho S, Jeong GJ, Kim BS, Hwang NS, Park TH. Lineage Specific Differentiation of Magnetic Nanoparticle-Based Size Controlled Human Embryoid Body. ACS Biomater Sci Eng 2017; 3:1719-1729. [DOI: 10.1021/acsbiomaterials.7b00141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Boram Son
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Jeong Ah Kim
- Biomedical
Omics Group, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea
| | - Sungwoo Cho
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Gun-Jae Jeong
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Byung Soo Kim
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Nathaniel S. Hwang
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Tai Hyun Park
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| |
Collapse
|
197
|
Kugler J, Huhse B, Tralau T, Luch A. Embryonic stem cells and the next generation of developmental toxicity testing. Expert Opin Drug Metab Toxicol 2017; 13:833-841. [PMID: 28675072 DOI: 10.1080/17425255.2017.1351548] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The advent of stem cell technology has seen the establishment of embryonic stem cells (ESCs) as molecular model systems and screening tools. Although ESCs are nowadays widely used in research, regulatory implementation for developmental toxicity testing is pending. Areas Covered: This review evaluates the performance of current ESC, including human (h)ESC testing systems, trying to elucidate their potential for developmental toxicity testing. It shall discuss defining parameters and mechanisms, their relevance and contemplate what can realistically be expected. Crucially this includes the question of how to ascertain the quality of currently employed cell lines and tests based thereon. Finally, the use of hESCs will raise ethical concerns which should be addressed early on. Expert Opinion: While the suitability of (h)ESCs as tools for research and development goes undisputed, any routine use for developmental toxicity testing currently still seems premature. The reasons for this comprise inherent biological deficiencies as well as cell line quality and system validation. Overcoming these issues will require collaboration of scientists, test developers and regulators. Also, validation needs to be made worthwhile for academia. Finally we have to continuously rethink existing strategies, making room for improved testing and innovative approaches.
Collapse
Affiliation(s)
- Josephine Kugler
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| | - Bettina Huhse
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| | - Tewes Tralau
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| | - Andreas Luch
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| |
Collapse
|
198
|
Li Y, Wen Y, Green M, Cabral EK, Wani P, Zhang F, Wei Y, Baer TM, Chen B. Cell sex affects extracellular matrix protein expression and proliferation of smooth muscle progenitor cells derived from human pluripotent stem cells. Stem Cell Res Ther 2017; 8:156. [PMID: 28676082 PMCID: PMC5496346 DOI: 10.1186/s13287-017-0606-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/01/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022] Open
Abstract
Background Smooth muscle progenitor cells (pSMCs) differentiated from human pluripotent stem cells (hPSCs) hold great promise for treating diseases or degenerative conditions involving smooth muscle pathologies. However, the therapeutic potential of pSMCs derived from men and women may be very different. Cell sex can exert a profound impact on the differentiation process of stem cells into somatic cells. In spite of advances in translation of stem cell technologies, the role of cell sex and the effect of sex hormones on the differentiation towards mesenchymal lineage pSMCs remain largely unexplored. Methods Using a standard differentiation protocol, two human embryonic stem cell lines (one male line and one female line) and three induced pluripotent stem cell lines (one male line and two female lines) were differentiated into pSMCs. We examined differences in the differentiation of male and female hPSCs into pSMCs, and investigated the effect of 17β-estradiol (E2) on the extracellular matrix (ECM) metabolisms and cell proliferation rates of the pSMCs. Statistical analyses were performed by using Student’s t test or two-way ANOVA, p < 0.05. Results Male and female hPSCs had similar differentiation efficiencies and generated morphologically comparable pSMCs under a standard differentiation protocol, but the derived pSMCs showed sex differences in expression of ECM proteins, such as MMP-2 and TIMP-1, and cell proliferation rates. E2 treatment induced the expression of myogenic gene markers and suppressed ECM degradation activities through reduction of MMP activity and increased expression of TIMP-1 in female pSMCs, but not in male pSMCs. Conclusions hPSC-derived pSMCs from different sexes show differential expression of ECM proteins and proliferation rates. Estrogen appears to promote maturation and ECM protein expression in female pSMCs, but not in male pSMCs. These data suggest that intrinsic cell-sex differences may influence progenitor cell biology. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0606-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA.,Department of Obstetrics/Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yan Wen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA.
| | - Morgaine Green
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Elise K Cabral
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Prachi Wani
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Fan Zhang
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Yi Wei
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Thomas M Baer
- Stanford Photonics Research Center, Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Bertha Chen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| |
Collapse
|
199
|
Zimmerlin L, Park TS, Zambidis ET. Capturing Human Naïve Pluripotency in the Embryo and in the Dish. Stem Cells Dev 2017; 26:1141-1161. [PMID: 28537488 DOI: 10.1089/scd.2017.0055] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although human embryonic stem cells (hESCs) were first derived almost 20 years ago, it was only recently acknowledged that they share closer molecular and functional identity to postimplantation lineage-primed murine epiblast stem cells than to naïve preimplantation inner cell mass-derived mouse ESCs (mESCs). A myriad of transcriptional, epigenetic, biochemical, and metabolic attributes have now been described that distinguish naïve and primed pluripotent states in both rodents and humans. Conventional hESCs and human induced pluripotent stem cells (hiPSCs) appear to lack many of the defining hallmarks of naïve mESCs. These include important features of the naïve ground state murine epiblast, such as an open epigenetic architecture, reduced lineage-primed gene expression, and chimera and germline competence following injection into a recipient blastocyst-stage embryo. Several transgenic and chemical methods were recently reported that appear to revert conventional human PSCs to mESC-like ground states. However, it remains unclear if subtle deviations in global transcription, cell signaling dependencies, and extent of epigenetic/metabolic shifts in these various human naïve-reverted pluripotent states represent true functional differences or alternatively the existence of distinct human pluripotent states along a spectrum. In this study, we review the current understanding and developmental features of various human pluripotency-associated phenotypes and discuss potential biological mechanisms that may support stable maintenance of an authentic epiblast-like ground state of human pluripotency.
Collapse
Affiliation(s)
- Ludovic Zimmerlin
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| | - Tea Soon Park
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| | - Elias T Zambidis
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| |
Collapse
|
200
|
Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc 2017. [DOI: 10.1111/brv.12349] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sara Al-Khawaga
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Bushra Memon
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Alexandra E. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine; University of California; Los Angeles CA 90095 U.S.A
| | - Shahrad Taheri
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Abdul B. Abou-Samra
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Essam M. Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|