1
|
Gao P, Liu S, Wang X, Ikeya M. Dental applications of induced pluripotent stem cells and their derivatives. JAPANESE DENTAL SCIENCE REVIEW 2022; 58:162-171. [PMID: 35516907 PMCID: PMC9065891 DOI: 10.1016/j.jdsr.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 11/26/2022] Open
Abstract
Periodontal tissue regeneration is the ideal tactic for treating periodontitis. Tooth regeneration is the potential strategy to restore the lost teeth. With infinite self-renewal, broad differentiation potential, and less ethical issues than embryonic stem cells, induced pluripotent stem cells (iPSCs) are promising cell resource for periodontal and tooth regeneration. This review summarized the optimized technologies of generating iPSC lines and application of iPSC derivatives, which reduce the risk of tumorigenicity. Given that iPSCs may have epigenetic memory from the donor tissue and tend to differentiate into lineages along with the donor cells, iPSCs derived from dental tissues may benefit for personalized dental application. Neural crest cells (NCCs) and mesenchymal stem or stomal cells (MSCs) are lineage-specific progenitor cells derived from iPSCs and can differentiate into multilineage cell types. This review introduced the updated technologies of inducing iPSC-derived NCCs and iPSC-derived MSCs and their application in periodontal and tooth regeneration. Given the complexity of periodontal tissues and teeth, it is crucial to elucidate the integrated mechanisms of all constitutive cells and the spatio-temporal interactions among them to generate structural periodontal tissues and functional teeth. Thus, more sophisticated studies in vitro and in vivo and even preclinical investigations need to be conducted.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of General and Emergency Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shan Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Division of Oral Ecology and Biochemistry, Oral Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Xiaoyi Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Vandana JJ, Lacko LA, Chen S. Phenotypic technologies in stem cell biology. Cell Chem Biol 2021; 28:257-270. [PMID: 33651977 DOI: 10.1016/j.chembiol.2021.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
The high-throughput phenotypic screen (HTPS) has become an emerging technology to discover synthetic small molecules that regulate stem cell fates. Here, we review the application of HTPS to identify small molecules controlling stem cell renewal, reprogramming, differentiation, and lineage conversion. Moreover, we discuss the use of HTPS to discover small molecules/polymers mimicking the stem cell extracellular niche. Furthermore, HTPSs have been applied on whole-animal models to identify small molecules regulating stem cell renewal or differentiation in vivo. Finally, we discuss the examples of the utilization of HTPS in stem cell-based disease modeling, as well as in the discovery of novel drug candidates for cancer, diabetes, and infectious diseases. Overall, HTPSs have provided many powerful tools for the stem cell field, which not only facilitate the generation of functional cells/tissues for replacement therapy, disease modeling, and drug screening, but also help dissect molecular mechanisms regulating physiological and pathological processes.
Collapse
Affiliation(s)
- J Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Tri-Institutional PhD Program in Chemical Biology, Weill Cornell Medicine, The Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauretta A Lacko
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
3
|
A Concise Review on Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Personalized Regenerative Medicine. Stem Cell Rev Rep 2020; 17:748-776. [PMID: 33098306 DOI: 10.1007/s12015-020-10061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
The induced pluripotent stem cells (iPSCs) are derived from somatic cells by using reprogramming factors such as Oct4, Sox2, Klf4, and c-Myc (OSKM) or Oct4, Sox2, Nanog and Lin28 (OSNL). They resemble embryonic stem cells (ESCs) and have the ability to differentiate into cell lineage of all three germ-layer, including cardiomyocytes (CMs). The CMs can be generated from iPSCs by inducing embryoid bodies (EBs) formation and treatment with activin A, bone morphogenic protein 4 (BMP4), and inhibitors of Wnt signaling. However, these iPSC-derived CMs are a heterogeneous population of cells and require purification and maturation to mimic the in vivo CMs. The matured CMs can be used for various therapeutic purposes in regenerative medicine by cardiomyoplasty or through the development of tissue-engineered cardiac patches. In recent years, significant advancements have been made in the isolation of iPSC and their differentiation, purification, and maturation into clinically usable CMs. Newer small molecules have also been identified to substitute the reprogramming factors for iPSC generation as well as for direct differentiation of somatic cells into CMs without an intermediary pluripotent state. This review provides a concise update on the generation of iPSC-derived CMs and their application in personalized cardiac regenerative medicine. It also discusses the current limitations and challenges in the application of iPSC-derived CMs. Graphical abstract.
Collapse
|
4
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
5
|
Borgohain MP, Haridhasapavalan KK, Dey C, Adhikari P, Thummer RP. An Insight into DNA-free Reprogramming Approaches to Generate Integration-free Induced Pluripotent Stem Cells for Prospective Biomedical Applications. Stem Cell Rev Rep 2020; 15:286-313. [PMID: 30417242 DOI: 10.1007/s12015-018-9861-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than a decade ago, a pioneering study reported generation of induced Pluripotent Stem Cells (iPSCs) by ectopic expression of a cocktail of reprogramming factors in fibroblasts. This study has revolutionized stem cell research and has garnered immense interest from the scientific community globally. iPSCs hold tremendous potential for understanding human developmental biology, disease modeling, drug screening and discovery, and personalized cell-based therapeutic applications. The seminal study identified Oct4, Sox2, Klf4 and c-Myc as a potent combination of genes to induce reprogramming. Subsequently, various reprogramming factors were identified by numerous groups. Most of these studies have used integrating viral vectors to overexpress reprogramming factors in somatic cells to derive iPSCs. However, these techniques restrict the clinical applicability of these cells as they may alter the genome due to random viral integration resulting in insertional mutagenesis and tumorigenicity. To circumvent this issue, alternative integration-free reprogramming approaches are continuously developed that eliminate the risk of genomic modifications and improve the prospects of iPSCs from lab to clinic. These methods establish that integration of transgenes into the genome is not essential to induce pluripotency in somatic cells. This review provides a comprehensive overview of the most promising DNA-free reprogramming techniques that have the potential to derive integration-free iPSCs without genomic manipulation, such as sendai virus, recombinant proteins, microRNAs, synthetic messenger RNA and small molecules. The understanding of these approaches shall pave a way for the generation of clinical-grade iPSCs. Subsequently, these iPSCs can be differentiated into desired cell type(s) for various biomedical applications.
Collapse
Affiliation(s)
- Manash P Borgohain
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Chandrima Dey
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Poulomi Adhikari
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
6
|
Yavarpour-Bali H, Ghasemi-Kasman M, Shojaei A. Direct reprogramming of terminally differentiated cells into neurons: A novel and promising strategy for Alzheimer's disease treatment. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109820. [PMID: 31743695 DOI: 10.1016/j.pnpbp.2019.109820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 01/17/2023]
Abstract
Glial activation is a common pathological process of the central nervous system (CNS) in disorders such as Alzheimer's disease (AD). Several approaches have been used to reduce the number of activated astrocytes and microglia in damaged areas. In recent years, various kinds of fully differentiated cells have been successfully reprogrammed to a desired type of cell in lesion areas. Interestingly, internal glial cells, including astrocytes and NG2 positive cells, were efficiently converted to neuroblasts and neurons by overexpression of some transcription factors (TFs) or microRNAs (miRNAs). Notably, some specific subtypes of neurons have been achieved by in vivo reprogramming and the resulting neurons were successfully integrated into local neuronal circuits. Furthermore, somatic cells from AD patients have been converted to functional neurons. Although direct reprogramming of a patient's own internal cells has revolutionized regenerative medicine, but there are some major obstacles that should be examined before using these induced cells in clinical therapies. In the present review article, we aim to discuss the current studies on in vitro and in vivo reprogramming of somatic cells to neurons using TFs, miRNAs or small molecules in healthy and AD patients.
Collapse
Affiliation(s)
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Amir Shojaei
- Department of Physiology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
7
|
Protein Kinases and Their Inhibitors in Pluripotent Stem Cell Fate Regulation. Stem Cells Int 2019; 2019:1569740. [PMID: 31428157 PMCID: PMC6681599 DOI: 10.1155/2019/1569740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/31/2019] [Accepted: 06/16/2019] [Indexed: 12/25/2022] Open
Abstract
Protein kinases modulate the reversible postmodifications of substrate proteins to their phosphorylated forms as an essential process in regulating intracellular signaling transduction cascades. Moreover, phosphorylation has recently been shown to tightly control the regulatory network of kinases responsible for the induction and maintenance of pluripotency, defined as the particular ability to differentiate pluripotent stem cells (PSCs) into every cell type in the adult body. In particular, emerging evidence indicates that the balance between the self-renewal and differentiation of PSCs is regulated by the small molecules that modulate kinase signaling pathways. Furthermore, new reprogramming technologies have been developed using kinase modulators, which have provided novel insight of the mechanisms underlying the kinase regulatory networks involved in the generation of induced pluripotent stem cells (iPSCs). In this review, we highlight the recent progress made in defining the roles of protein kinase signaling pathways and their small molecule modulators in regulating the pluripotent states, self-renewal, reprogramming process, and lineage differentiation of PSCs.
Collapse
|
8
|
Abu-Dawud R, Graffmann N, Ferber S, Wruck W, Adjaye J. Pluripotent stem cells: induction and self-renewal. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0213. [PMID: 29786549 DOI: 10.1098/rstb.2017.0213] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2017] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cells (PSCs) lie at the heart of modern regenerative medicine due to their properties of unlimited self-renewal in vitro and their ability to differentiate into cell types representative of the three embryonic germ layers-mesoderm, ectoderm and endoderm. The derivation of induced PSCs bypasses ethical concerns associated with the use of human embryonic stem cells and also enables personalized cell-based therapies. To exploit their regenerative potential, it is essential to have a firm understanding of the molecular processes associated with their induction from somatic cells. This understanding serves two purposes: first, to enable efficient, reliable and cost-effective production of excellent quality induced PSCs and, second, to enable the derivation of safe, good manufacturing practice-grade transplantable donor cells. Here, we review the reprogramming process of somatic cells into induced PSCs and associated mechanisms with emphasis on self-renewal, epigenetic control, mitochondrial bioenergetics, sub-states of pluripotency, naive ground state, naive and primed. A meta-analysis identified genes expressed exclusively in the inner cell mass and in the naive but not in the primed pluripotent state. We propose these as additional biomarkers defining naive PSCs.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- R Abu-Dawud
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Zahrawi Street, Riyadh 11211, Saudi Arabia
| | - N Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - S Ferber
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - W Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - J Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| |
Collapse
|
9
|
Chen X, Li J, Huang Y, Liu P, Fan Y. Insoluble Microenvironment Facilitating the Generation and Maintenance of Pluripotency. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:267-278. [PMID: 29327674 DOI: 10.1089/ten.teb.2017.0415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold enormous potential as a tool to generate cells for tissue engineering and regenerative medicine. Since the initial report of iPSCs in 2006, many different methods have been developed to enhance the safety and efficiency of this technology. Recent studies indicate that the extracellular signals can promote the production of iPSCs, and even replace the Yamanaka factors. Noticeably, abundant evidences suggest that the insoluble microenvironment, including the culture substrate and neighboring cells, directly regulates the expression of core pluripotency genes and the epigenetic modification of the chromatins, hence, impacts the reprogramming dynamics. These studies provide new strategies for developing safer and more efficient method for iPSC generation. In this review, we examine the publications addressing the insoluble extracellular microenvironment that boosts iPSC generation and self-renewal. We also discuss cell adhesion-mediated molecular mechanisms, through which the insoluble extracellular cues interplay with reprogramming.
Collapse
Affiliation(s)
- Xiaofang Chen
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Jiaqi Li
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
| | - Yan Huang
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Peng Liu
- 3 Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University , Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
- 4 National Research Center for Rehabilitation Technical Aids , Beijing, China
| |
Collapse
|
10
|
Choi B, Kim D, Han I, Lee SH. Microenvironmental Regulation of Stem Cell Behavior Through Biochemical and Biophysical Stimulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:147-160. [PMID: 30471031 DOI: 10.1007/978-981-13-0445-3_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stem cells proliferate by undergoing self-renewal and differentiate into multiple cell lineages in response to biochemical and biophysical stimuli. Various biochemical cues such as growth factors, nucleic acids, chemical reagents, and small molecules have been used to induce stem cell differentiation or reprogramming or to maintain their pluripotency. Moreover, biophysical cues such as matrix stiffness, substrate topography, and external stress and strain play a major role in modulating stem cell behavior. In this chapter, we have summarized microenvironmental regulation of stem cell behavior through biochemical and biophysical stimulation.
Collapse
Affiliation(s)
- Bogyu Choi
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Deogil Kim
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, South Korea
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Gyeonggi-do, South Korea.
| |
Collapse
|
11
|
Replacing reprogramming factors with antibodies selected from combinatorial antibody libraries. Nat Biotechnol 2017; 35:960-968. [PMID: 28892074 DOI: 10.1038/nbt.3963] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 08/16/2017] [Indexed: 01/12/2023]
Abstract
The reprogramming of differentiated cells into induced pluripotent stem cells (iPSCs) is usually achieved by exogenous induction of transcription by factors acting in the nucleus. In contrast, during development, signaling pathways initiated at the membrane induce differentiation. The central idea of this study is to identify antibodies that can catalyze cellular de-differentiation and nuclear reprogramming by acting at the cell surface. We screen a lentiviral library encoding ∼100 million secreted and membrane-bound single-chain antibodies and identify antibodies that can replace either Sox2 and Myc (c-Myc) or Oct4 during reprogramming of mouse embryonic fibroblasts into iPSCs. We show that one Sox2-replacing antibody antagonizes the membrane-associated protein Basp1, thereby de-repressing nuclear factors WT1, Esrrb and Lin28a (Lin28) independent of Sox2. By manipulating this pathway, we identify three methods to generate iPSCs. Our results establish unbiased selection from autocrine combinatorial antibody libraries as a robust method to discover new biologics and uncover membrane-to-nucleus signaling pathways that regulate pluripotency and cell fate.
Collapse
|
12
|
Baranek M, Belter A, Naskręt-Barciszewska MZ, Stobiecki M, Markiewicz WT, Barciszewski J. Effect of small molecules on cell reprogramming. MOLECULAR BIOSYSTEMS 2017; 13:277-313. [PMID: 27918060 DOI: 10.1039/c6mb00595k] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The essential idea of regenerative medicine is to fix or replace tissues or organs with alive and patient-specific implants. Pluripotent stem cells are able to indefinitely self-renew and differentiate into all cell types of the body which makes them a potent substantial player in regenerative medicine. The easily accessible source of induced pluripotent stem cells may allow obtaining and cultivating tissues in vitro. Reprogramming refers to regression of mature cells to its initial pluripotent state. One of the approaches affecting pluripotency is the usage of low molecular mass compounds that can modulate enzymes and receptors leading to the formation of pluripotent stem cells (iPSCs). It would be great to assess the general character of such compounds and reveal their new derivatives or modifications to increase the cell reprogramming efficiency. Many improvements in the methods of pluripotency induction have been made by various groups in order to limit the immunogenicity and tumorigenesis, increase the efficiency and accelerate the kinetics. Understanding the epigenetic changes during the cellular reprogramming process will extend the comprehension of stem cell biology and lead to potential therapeutic approaches. There are compounds which have been already proven to be or for now only putative inducers of the pluripotent state that may substitute for the classic reprogramming factors (Oct3/4, Sox2, Klf4, c-Myc) in order to improve the time and efficiency of pluripotency induction. The effect of small molecules on gene expression is dosage-dependent and their application concentration needs to be strictly determined. In this review we analysed the role of small molecules in modulations leading to pluripotency induction, thereby contributing to our understanding of stem cell biology and uncovering the major mechanisms involved in that process.
Collapse
Affiliation(s)
- M Baranek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - A Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Z Naskręt-Barciszewska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Stobiecki
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - W T Markiewicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - J Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| |
Collapse
|
13
|
Boda E, Nato G, Buffo A. Emerging pharmacological approaches to promote neurogenesis from endogenous glial cells. Biochem Pharmacol 2017. [PMID: 28647491 DOI: 10.1016/j.bcp.2017.06.129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are emerging as leading contributors to the global disease burden. While some drug-based approaches have been designed to limit or prevent neuronal loss following acute damage or chronic neurodegeneration, regeneration of functional neurons in the adult Central Nervous System (CNS) still remains an unmet need. In this context, the exploitation of endogenous cell sources has recently gained an unprecedented attention, thanks to the demonstration that, in some CNS regions or under specific circumstances, glial cells can activate spontaneous neurogenesis or can be instructed to produce neurons in the adult mammalian CNS parenchyma. This field of research has greatly advanced in the last years and identified interesting molecular and cellular mechanisms guiding the neurogenic activation/conversion of glia. In this review, we summarize the evolution of the research devoted to understand how resident glia can be directed to produce neurons. We paid particular attention to pharmacologically-relevant approaches exploiting the modulation of niche-associated factors and the application of selected small molecules.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy.
| | - Giulia Nato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| |
Collapse
|
14
|
Davis TL, Rebay I. Master regulators in development: Views from the Drosophila retinal determination and mammalian pluripotency gene networks. Dev Biol 2016; 421:93-107. [PMID: 27979656 DOI: 10.1016/j.ydbio.2016.12.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/03/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023]
Abstract
Among the mechanisms that steer cells to their correct fate during development, master regulatory networks are unique in their sufficiency to trigger a developmental program outside of its normal context. In this review we discuss the key features that underlie master regulatory potency during normal and ectopic development, focusing on two examples, the retinal determination gene network (RDGN) that directs eye development in the fruit fly and the pluripotency gene network (PGN) that maintains cell fate competency in the early mammalian embryo. In addition to the hierarchical transcriptional activation, extensive positive transcriptional feedback, and cooperative protein-protein interactions that enable master regulators to override competing cellular programs, recent evidence suggests that network topology must also be dynamic, with extensive rewiring of the interactions and feedback loops required to navigate the correct sequence of developmental transitions to reach a final fate. By synthesizing the in vivo evidence provided by the RDGN with the extensive mechanistic insight gleaned from the PGN, we highlight the unique regulatory capabilities that continual reorganization into new hierarchies confers on master control networks. We suggest that deeper understanding of such dynamics should be a priority, as accurate spatiotemporal remodeling of network topology will undoubtedly be essential for successful stem cell based therapeutic efforts.
Collapse
Affiliation(s)
- Trevor L Davis
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Ilaria Rebay
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA; Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
15
|
Small molecules increase direct neural conversion of human fibroblasts. Sci Rep 2016; 6:38290. [PMID: 27917895 PMCID: PMC5137010 DOI: 10.1038/srep38290] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/07/2016] [Indexed: 12/24/2022] Open
Abstract
The generation of human induced neurons (hiNs) via exogenous delivery of neural transcription factors represents a novel technique to obtain disease and patient specific neurons. These cells have the potential to be used for disease modeling, diagnostics and drug screening, and also to be further developed for brain repair. In the present study, we utilized hiNs to develop an unbiased screening assay for small molecules that increase the conversion efficiency. Using this assay, we screened 307 compounds from five annotated libraries and identified six compounds that were very potent in potentiating the reprogramming process. When combined in an optimal combination and dose, these compounds increased the reprogramming efficiency of human fibroblasts more than 6-fold. Global gene expression and CellNet analysis at different timepoints during the reprogramming process revealed that neuron-specific genes and gene regulatory networks (GRNs) became progressively more activated while converting cells shut down fibroblast-specific GRNs. Further bioinformatics analysis revealed that the addition of the six compound resulted in the accelerated upregulation of a subset of neuronal genes, and also increased expression of genes associated with transcriptional activity and mediation of cellular stress response.
Collapse
|
16
|
Zhang Q, Chen W, Tan S, Lin T. Stem Cells for Modeling and Therapy of Parkinson's Disease. Hum Gene Ther 2016; 28:85-98. [PMID: 27762639 DOI: 10.1089/hum.2016.116] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disease after Alzheimer's disease, which is characterized by a low level of dopamine being expressing in the striatum and a deterioration of dopaminergic neurons (DAn) in the substantia nigra pars compacta. Generation of PD-derived DAn, including differentiation of human embryonic stem cells, human neural stem cells, human-induced pluripotent stem cells, and direct reprogramming, provides an ideal tool to model PD, creating the possibility of mimicking key essential pathological processes and charactering single-cell changes in vitro. Furthermore, thanks to the understanding of molecular neuropathogenesis of PD and new advances in stem-cell technology, it is anticipated that optimal functionally transplanted DAn with targeted correction and transgene-free insertion will be generated for use in cell transplantation. This review elucidates stem-cell technology for modeling PD and offering desired safe cell resources for cell transplantation therapy.
Collapse
Affiliation(s)
- Qingxi Zhang
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Wanling Chen
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Sheng Tan
- 2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Tongxiang Lin
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,3 Stem Cell Research Center, Fujian Agriculture and Forestry University , Fuzhou, China
| |
Collapse
|
17
|
Xiao X, Li N, Zhang D, Yang B, Guo H, Li Y. Generation of Induced Pluripotent Stem Cells with Substitutes for Yamanaka's Four Transcription Factors. Cell Reprogram 2016; 18:281-297. [PMID: 27696909 DOI: 10.1089/cell.2016.0020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) share many characteristics with embryonic stem cells, but lack ethical controversy. They provide vast opportunities for disease modeling, pathogenesis understanding, therapeutic drug development, toxicology, organ synthesis, and treatment of degenerative disease. However, this procedure also has many potential challenges, including a slow generation time, low efficiency, partially reprogrammed colonies, as well as somatic coding mutations in the genome. Pioneered by Shinya Yamanaka's team in 2006, iPSCs were first generated by introducing four transcription factors: Oct 4, Sox 2, Klf 4, and c-Myc (OSKM). Of those factors, Klf 4 and c-Myc are oncogenes, which are potentially a tumor risk. Therefore, to avoid problems such as tumorigenesis and low throughput, one of the key strategies has been to use other methods, including members of the same subgroup of transcription factors, activators or inhibitors of signaling pathways, microRNAs, epigenetic modifiers, or even differentiation-associated factors, to functionally replace the reprogramming transcription factors. In this study, we will mainly focus on the advances in the generation of iPSCs with substitutes for OSKM. The identification and combination of novel proteins or chemicals, particularly small molecules, to induce pluripotency will provide useful tools to discover the molecular mechanisms governing reprogramming and ultimately lead to the development of new iPSC-based therapeutics for future clinical applications.
Collapse
Affiliation(s)
- Xiong Xiao
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China .,2 Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Nan Li
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China
| | - Dapeng Zhang
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China
| | - Bo Yang
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China
| | - Hongmei Guo
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China
| | - Yuemin Li
- 1 College of Animal Science and Technology, Southwest University , Chongqing, China
| |
Collapse
|
18
|
Wang B, Pfeiffer MJ, Drexler HCA, Fuellen G, Boiani M. Proteomic Analysis of Mouse Oocytes Identifies PRMT7 as a Reprogramming Factor that Replaces SOX2 in the Induction of Pluripotent Stem Cells. J Proteome Res 2016; 15:2407-21. [PMID: 27225728 DOI: 10.1021/acs.jproteome.5b01083] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The reprogramming process that leads to induced pluripotent stem cells (iPSCs) may benefit from adding oocyte factors to Yamanaka's reprogramming cocktail (OCT4, SOX2, KLF4, with or without MYC; OSK(M)). We previously searched for such facilitators of reprogramming (the reprogrammome) by applying label-free LC-MS/MS analysis to mouse oocytes, producing a catalog of 28 candidates that are (i) able to robustly access the cell nucleus and (ii) shared between mature mouse oocytes and pluripotent embryonic stem cells. In the present study, we hypothesized that our 28 reprogrammome candidates would also be (iii) abundant in mature oocytes, (iv) depleted after the oocyte-to-embryo transition, and (v) able to potentiate or replace the OSKM factors. Using LC-MS/MS and isotopic labeling methods, we found that the abundance profiles of the 28 proteins were below those of known oocyte-specific and housekeeping proteins. Of the 28 proteins, only arginine methyltransferase 7 (PRMT7) changed substantially during mouse embryogenesis and promoted the conversion of mouse fibroblasts into iPSCs. Specifically, PRMT7 replaced SOX2 in a factor-substitution assay, yielding iPSCs. These findings exemplify how proteomics can be used to prioritize the functional analysis of reprogrammome candidates. The LC-MS/MS data are available via ProteomeXchange with identifier PXD003093.
Collapse
Affiliation(s)
- Bingyuan Wang
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences , Beijing 100193, China
| | - Martin J Pfeiffer
- Max Planck Institute for Molecular Biomedicine , Röntgenstraße 20, 48149 Münster, Germany
| | - Hannes C A Drexler
- Max Planck Institute for Molecular Biomedicine , Bioanalytical Mass Spectrometry Facility, Röntgenstraße 20, 48149 Münster, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michele Boiani
- Max Planck Institute for Molecular Biomedicine , Röntgenstraße 20, 48149 Münster, Germany
| |
Collapse
|
19
|
Charaf L, Mahon FX, Lamrissi-Garcia I, Moranvillier I, Beliveau F, Cardinaud B, Dabernat S, de Verneuil H, Moreau-Gaudry F, Bedel A. Effect of tyrosine kinase inhibitors on stemness in normal and chronic myeloid leukemia cells. Leukemia 2016; 31:65-74. [PMID: 27220663 DOI: 10.1038/leu.2016.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022]
Abstract
Although tyrosine kinase inhibitors (TKIs) efficiently cure chronic myeloid leukemia (CML), they can fail to eradicate CML stem cells (CML-SCs). The mechanisms responsible for CML-SC survival need to be understood for designing therapies. Several previous studies suggest that TKIs could modulate CML-SC quiescence. Unfortunately, CML-SCs are insufficiently available. Induced pluripotent stem cells (iPSCs) offer a promising alternative. In this work, we used iPSCs derived from CML patients (Ph+). Ph+ iPSC clones expressed lower levels of stemness markers than normal iPSCs. BCR-ABL1 was found to be involved in stemness regulation and ERK1/2 to have a key role in the signaling pathway. TKIs unexpectedly promoted stemness marker expression in Ph+ iPSC clones. Imatinib also retained quiescence and induced stemness gene expression in CML-SCs. Our results suggest that TKIs might have a role in residual disease and confirm the need for a targeted therapy different from TKIs that could overcome the stemness-promoting effect caused by TKIs. Interestingly, a similar pro-stemness effect was observed in normal iPSCs and hematopoietic SCs. These findings could help to explain CML resistance mechanisms and the teratogenic side-effects of TKIs in embryonic cells.
Collapse
Affiliation(s)
- L Charaf
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France
| | - F-X Mahon
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Pôle de Biologie et Pathologie CHU Bordeaux, Bordeaux, France.,Institut Bergonie, SIRIC BRIO, Bordeaux, France
| | - I Lamrissi-Garcia
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France
| | - I Moranvillier
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France
| | - F Beliveau
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France
| | - B Cardinaud
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Institut Polytechnique de Bordeaux, Talence, France
| | - S Dabernat
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France.,Pôle de Biologie et Pathologie CHU Bordeaux, Bordeaux, France
| | - H de Verneuil
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France.,Pôle de Biologie et Pathologie CHU Bordeaux, Bordeaux, France
| | - F Moreau-Gaudry
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France.,Pôle de Biologie et Pathologie CHU Bordeaux, Bordeaux, France
| | - A Bedel
- Inserm U1035, Biothérapies des Maladies Génétiques et Cancers, Bordeaux, France.,Université de Bordeaux, FR TransBiomed, Bordeaux, France.,Laboratoire d'Excellence GR-Ex, Bordeaux, France.,Pôle de Biologie et Pathologie CHU Bordeaux, Bordeaux, France
| |
Collapse
|
20
|
Pluripotent Stem Cells: Current Understanding and Future Directions. Stem Cells Int 2015; 2016:9451492. [PMID: 26798367 PMCID: PMC4699068 DOI: 10.1155/2016/9451492] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells have the ability to undergo self-renewal and to give rise to all cells of the tissues of the body. However, this definition has been recently complicated by the existence of distinct cellular states that display these features. Here, we provide a detailed overview of the family of pluripotent cell lines derived from early mouse and human embryos and compare them with induced pluripotent stem cells. Shared and distinct features of these cells are reported as additional hallmark of pluripotency, offering a comprehensive scenario of pluripotent stem cells.
Collapse
|
21
|
Anwar MA, Kim S, Choi S. The triumph of chemically enhanced cellular reprogramming: a patent review. Expert Opin Ther Pat 2015; 26:265-80. [PMID: 26593376 DOI: 10.1517/13543776.2016.1118058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The revolutionary discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka has exposed science to new horizons. However, genetic modifications render reprogrammed cells unstable; for that reason, non-genetic modification approaches are actively under investigation. Among these, the use of small molecules is safe, and these molecules minimally affect the genome. Although iPSCs are ready for clinical trials there are many caveats hindering successful therapy, and small molecules are the best alternative to overcome those caveats. AREAS COVERED Small molecules are playing an active role in generating and improving the quality of iPSCs. In this review, we will highlight the imperative role of small molecules in accelerating the successful translation of basic research into clinical use. Particularly, those ligands that replace the need for reprogramming factors will be discussed. EXPERT OPINION Stem cell research is promising for harvesting medical benefits in near future. The invention of new techniques, mechanisms elucidation, and identification of novel compounds for stem cell creation has certainly established a solid foundation for regenerative medicine. This is the beginning of a new era for the cure of most disabling diseases, and small molecules will have a definite role in successful therapeutic use of iPSCs.
Collapse
Affiliation(s)
- Muhammad Ayaz Anwar
- a Department of Molecular Science and Technology , Ajou University , Suwon , South Korea
| | - Songmee Kim
- a Department of Molecular Science and Technology , Ajou University , Suwon , South Korea
| | - Sangdun Choi
- a Department of Molecular Science and Technology , Ajou University , Suwon , South Korea
| |
Collapse
|
22
|
González F, Huangfu D. Mechanisms underlying the formation of induced pluripotent stem cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:39-65. [PMID: 26383234 DOI: 10.1002/wdev.206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/13/2015] [Accepted: 07/21/2015] [Indexed: 12/19/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer unique opportunities for studying human biology, modeling diseases, and therapeutic applications. The simplest approach so far to generate human PSC lines is through reprogramming of somatic cells from an individual by defined factors, referred to simply as reprogramming. Reprogramming circumvents the ethical controversies associated with human embryonic stem cells (hESCs) and nuclear transfer hESCs (nt-hESCs), and the resulting induced pluripotent stem cells (hiPSCs) retain the same basic genetic makeup as the somatic cell used for reprogramming. Since the first report of iPSCs by Takahashi and Yamanaka (Cell 2006, 126:663-676), the molecular mechanisms of reprogramming have been extensively investigated. A better mechanistic understanding of reprogramming is fundamental not only to iPSC biology and improving the quality of iPSCs for therapeutic use, but also to our understanding of the molecular basis of cell identity, pluripotency, and plasticity. Here, we summarize the genetic, epigenetic, and cellular events during reprogramming, and the roles of various factors identified thus far in the reprogramming process. WIREs Dev Biol 2016, 5:39-65. doi: 10.1002/wdev.206 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Federico González
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY, USA
| |
Collapse
|
23
|
Reprogramming with Small Molecules instead of Exogenous Transcription Factors. Stem Cells Int 2015; 2015:794632. [PMID: 25922608 PMCID: PMC4397468 DOI: 10.1155/2015/794632] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 03/03/2015] [Accepted: 03/09/2015] [Indexed: 12/31/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) could be employed in the creation of patient-specific stem cells, which could subsequently be used in various basic and clinical applications. However, current iPSC methodologies present significant hidden risks with respect to genetic mutations and abnormal expression which are a barrier in realizing the full potential of iPSCs. A chemical approach is thought to be a promising strategy for safety and efficiency of iPSC generation. Many small molecules have been identified that can be used in place of exogenous transcription factors and significantly improve iPSC reprogramming efficiency and quality. Recent studies have shown that the use of small molecules results in the generation of chemically induced pluripotent stem cells from mouse embryonic fibroblast cells. These studies might lead to new areas of stem cell research and medical applications, not only human iPSC by chemicals alone, but also safe generation of somatic stem cells for cell based clinical trials and other researches. In this paper, we have reviewed the recent advances in small molecule approaches for the generation of iPSCs.
Collapse
|
24
|
Rony IK, Baten A, Bloomfield JA, Islam ME, Billah MM, Islam KD. Inducing pluripotency in vitro: recent advances and highlights in induced pluripotent stem cells generation and pluripotency reprogramming. Cell Prolif 2015; 48:140-56. [PMID: 25643745 DOI: 10.1111/cpr.12162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/05/2014] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are considered patient-specific counterparts of embryonic stem cells as they originate from somatic cells after forced expression of pluripotency reprogramming factors Oct4, Sox2, Klf4 and c-Myc. iPSCs offer unprecedented opportunity for personalized cell therapies in regenerative medicine. In recent years, iPSC technology has undergone substantial improvement to overcome slow and inefficient reprogramming protocols, and to ensure clinical-grade iPSCs and their functional derivatives. Recent developments in iPSC technology include better reprogramming methods employing novel delivery systems such as non-integrating viral and non-viral vectors, and characterization of alternative reprogramming factors. Concurrently, small chemical molecules (inhibitors of specific signalling or epigenetic regulators) have become crucial to iPSC reprogramming; they have the ability to replace putative reprogramming factors and boost reprogramming processes. Moreover, common dietary supplements, such as vitamin C and antioxidants, when introduced into reprogramming media, have been found to improve genomic and epigenomic profiles of iPSCs. In this article, we review the most recent advances in the iPSC field and potent application of iPSCs, in terms of cell therapy and tissue engineering.
Collapse
Affiliation(s)
- I K Rony
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | | | | | | | | | | |
Collapse
|
25
|
Li M, Li L, Zhang J, Verma V, Liu Q, Shi D, Huang B. An Insight on Small Molecule Induced Foot-Print Free Naive Pluripotent Stem Cells in Livestock. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/scd.2015.51001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Generation of pluripotent stem cells without the use of genetic material. J Transl Med 2015; 95:26-42. [PMID: 25365202 DOI: 10.1038/labinvest.2014.132] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/25/2014] [Accepted: 07/25/2014] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) provide a platform to obtain patient-specific cells for use as a cell source in regenerative medicine. Although iPSCs do not have the ethical concerns of embryonic stem cells, iPSCs have not been widely used in clinical applications, as they are generated by gene transduction. Recently, iPSCs have been generated without the use of genetic material. For example, protein-induced PSCs and chemically induced PSCs have been generated by the use of small and large (protein) molecules. Several epigenetic characteristics are important for cell differentiation; therefore, several small-molecule inhibitors of epigenetic-modifying enzymes, such as DNA methyltransferases, histone deacetylases, histone methyltransferases, and histone demethylases, are potential candidates for the reprogramming of somatic cells into iPSCs. In this review, we discuss what types of small chemical or large (protein) molecules could be used to replace the viral transduction of genes and/or genetic reprogramming to obtain human iPSCs.
Collapse
|
27
|
Li J, Song W, Pan G, Zhou J. Advances in understanding the cell types and approaches used for generating induced pluripotent stem cells. J Hematol Oncol 2014; 7:50. [PMID: 25037625 PMCID: PMC4445637 DOI: 10.1186/s13045-014-0050-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/01/2014] [Indexed: 02/06/2023] Open
Abstract
Successfully reprogramming somatic cells to a pluripotent state generates induced pluripotent stem (iPS) cells (or iPSCs), which have extensive self-renewal capacity like embryonic stem cells (ESCs). iPSCs can also generate daughter cells that can further undergo differentiation into various lineages or terminally differentiate to reach their final functional state. The discovery of how to produce iPSCs opened a new field of stem cell research with both intellectual and therapeutic benefits. The huge potential implications of disease-specific or patient-specific iPSCs have impelled scientists to solve problems hindering their applications in clinical medicine, especially the issues of convenience and safety. To determine the range of tissue types amenable to reprogramming as well as their particular characteristics, cells from three embryonic germ layers have been assessed, and the advantages that some tissue origins have over fibroblast origins concerning efficiency and accessibility have been elucidated. To provide safe iPSCs in an efficient and convenient way, the delivery systems and combinations of inducing factors as well as the chemicals used to generate iPSCs have also been significantly improved in addition to the efforts on finding better donor cells. Currently, iPSCs can be generated without c-Myc and Klf4 oncogenes, and non-viral delivery integration-free chemically mediated reprogramming methods have been successfully employed with relatively satisfactory efficiency. This paper will review recent advances in iPS technology by highlighting tissue origin and generation of iPSCs. The obstacles that need to be overcome for clinical applications of iPSCs are also discussed.
Collapse
Affiliation(s)
- Jun Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Weiqi Road, Jinan, 250021, P.R. China.
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Weiqi Road, Jinan, 250021, P.R. China.
| | - Guangjin Pan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, 190 Kaiyuan Avenue, Science Park, Guangzhou, 510530, P.R. China.
| | - Jun Zhou
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Weiqi Road, Jinan, 250021, P.R. China.
| |
Collapse
|
28
|
Feltes BC, Bonatto D. Combining small molecules for cell reprogramming through an interatomic analysis. MOLECULAR BIOSYSTEMS 2014; 9:2741-63. [PMID: 24056910 DOI: 10.1039/c3mb70159j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The knowledge available about the application and generation of induced pluripotent stem cells (iPSC) has grown since their discovery, and new techniques to enhance the reprogramming process have been described. Among the new approaches to induce iPSC that have gained great attention is the use of small molecules for reprogramming. The application of small molecules, unlike genetic manipulation, provides for control of the reprogramming process through the shifting of concentrations and the combination of different molecules. However, different researchers have reported the use of "reprogramming cocktails" with variable results and drug combinations. Thus, the proper combination of small molecules for successful and enhanced reprogramming is a matter for discussion. However, testing all potential drug combinations in different cell lineages is very costly and time-consuming. Therefore, in this article, we discuss the use of already employed molecules for iPSC generation, followed by the application of systems chemo-biology tools to create different data sets of protein-protein (PPI) and chemical-protein (CPI) interaction networks based on the knowledge of already used and new reprogramming cocktail combinations. We further analyzed the biological processes associated with PPI-CPI networks and provided new potential protein targets to be inhibited or expressed for stem cell reprogramming. In addition, we applied a new interference analysis to prospective targets that could negatively affect the classical pluripotency-associated factors (SOX2, NANOG, KLF4 and OCT4) and thus potentially improve reprogramming protocols.
Collapse
Affiliation(s)
- Bruno César Feltes
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500 - Prédio 43421 - Sala 219, Porto Alegre, Caixa Postal 15005, RS - Brazil.
| | | |
Collapse
|
29
|
Abstract
Small molecules that modulate stem cell fate and function offer significant opportunities that will allow the full realization of the therapeutic potential of stem cells. Rational design and screening for small molecules have identified useful compounds to probe fundamental mechanisms of stem cell self-renewal, differentiation, and reprogramming and have facilitated the development of cell-based therapies and therapeutic drugs targeting endogenous stem and progenitor cells for repair and regeneration. Here, we will discuss recent scientific and therapeutic progress, as well as new perspectives and future challenges for using chemical approaches in stem cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Wenlin Li
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | | | | | | |
Collapse
|
30
|
Jung DW, Kim WH, Williams DR. Reprogram or reboot: small molecule approaches for the production of induced pluripotent stem cells and direct cell reprogramming. ACS Chem Biol 2014; 9:80-95. [PMID: 24245936 DOI: 10.1021/cb400754f] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell transplantation is a potential therapy for regenerative medicine, which aims to restore tissues damaged by trauma, aging, and diseases. Since its conception in the late 1990s, chemical biology has provided powerful and diverse small molecule tools for modulating stem cell function. Embryonic stem cells could be an ideal source for transplantation, but ethical concerns restrict their development for cell therapy. The seminal advance of induced pluripotent stem cell (iPSC) technology provided an attractive alternative to human embryonic stem cells. However, iPSCs are not yet considered an ideal stem cell source, due to limitations associated with the reprogramming process and their potential tumorigenic behavior. This is an area of research where chemical biology has made a significant contribution to facilitate the efficient production of high quality iPSCs and elucidate the biological mechanisms governing their phenotype. In this review, we summarize these advances and discuss the latest progress in developing small molecule modulators. Moreover, we also review a new trend in stem cell research, which is the direct reprogramming of readily accessible cell types into clinically useful cells, such as neurons and cardiac cells. This is a research area where chemical biology is making a pivotal contribution and illustrates the many advantages of using small molecules in stem cell research.
Collapse
Affiliation(s)
- Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 1 Oryong-Dong, Buk-Gu, Gwangju 500-712, Republic of Korea
| | - Woong-Hee Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 1 Oryong-Dong, Buk-Gu, Gwangju 500-712, Republic of Korea
| | - Darren Reece Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 1 Oryong-Dong, Buk-Gu, Gwangju 500-712, Republic of Korea
| |
Collapse
|
31
|
Zhang X, Meyn MA, Smithgall TE. c-Yes tyrosine kinase is a potent suppressor of ES cell differentiation and antagonizes the actions of its closest phylogenetic relative, c-Src. ACS Chem Biol 2014; 9:139-46. [PMID: 23895624 DOI: 10.1021/cb400249b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Embryonic stem (ES) cells are derived from the inner cell mass of the blastocyst stage embryo and are characterized by self-renewal and pluripotency. Previous work has shown that Src-family tyrosine kinases display dynamic expression and activity changes during ES cell differentiation, suggesting distinct functions in the control of developmental fate. Here we used ES cells to test the hypothesis that c-Src and its closest phylogenetic relative, c-Yes, act in biological opposition despite their strong homology. Unlike c-Src, enforced expression of active c-Yes blocked ES cell differentiation to embryoid bodies by maintaining pluripotency gene expression. To explore the interplay of c-Src and c-Yes in ES cell differentiation, we engineered c-Src and c-Yes mutants that are resistant to A-419259, a potent pyrrolopyrimidine inhibitor of the Src kinase family. Previous studies have shown that A-419259 treatment blocks all Src-family kinase activity in ES cells, preventing differentiation while maintaining pluripotency. Expression of inhibitor-resistant c-Src but not c-Yes rescued the A-419259 differentiation block, resulting in a cell population with properties of both primitive ectoderm and endoderm. Remarkably, when inhibitor-resistant c-Src and c-Yes were expressed together in ES cells, c-Yes activity suppressed c-Src-mediated differentiation. These studies show that even closely related kinases such as c-Src and c-Yes have unique and opposing functions in the same cell type. Selective agonists or inhibitors of c-Src versus c-Yes activity may allow more precise pharmacological manipulation of ES cell fate and have broader applications in other biological systems that express multiple Src family members such as tumor cells.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania 15219, United States
| | - Malcolm A. Meyn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania 15219, United States
| | - Thomas E. Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania 15219, United States
| |
Collapse
|
32
|
Yuan Y, Hartland K, Boskovic Z, Wang Y, Walpita D, Lysy PA, Zhong C, Young DW, Kim YK, Tolliday NJ, Sokal EM, Schreiber SL, Wagner BK. A small-molecule inducer of PDX1 expression identified by high-throughput screening. ACTA ACUST UNITED AC 2013; 20:1513-22. [PMID: 24290880 DOI: 10.1016/j.chembiol.2013.10.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 01/05/2023]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1), a member of the homeodomain-containing transcription factor family, is a key transcription factor important for both pancreas development and mature β cell function. The ectopic overexpression of Pdx1, Neurog3, and MafA in mice reprograms acinar cells to insulin-producing cells. We developed a quantitative PCR-based gene expression assay to screen more than 60,000 compounds for expression of each of these genes in the human PANC-1 ductal carcinoma cell line. We identified BRD7552, which upregulated PDX1 expression in both primary human islets and ductal cells, and induced epigenetic changes in the PDX1 promoter consistent with transcriptional activation. Prolonged compound treatment induced both insulin mRNA and protein and also enhanced insulin expression induced by the three-gene combination. These results provide a proof of principle for identifying small molecules that induce expression of transcription factors to control cellular reprogramming.
Collapse
Affiliation(s)
- Yuan Yuan
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kate Hartland
- Chemical Biology Platform, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Zarko Boskovic
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Yikai Wang
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Deepika Walpita
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Philippe A Lysy
- Laboratory of Pediatric Hepatology and Cell Therapy, Catholic University of Leuven, Brussels 1200, Belgium
| | - Cheng Zhong
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Damian W Young
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Young-Kwon Kim
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nicola J Tolliday
- Chemical Biology Platform, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Etienne M Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Catholic University of Leuven, Brussels 1200, Belgium
| | - Stuart L Schreiber
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bridget K Wagner
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
33
|
The use of small molecules in somatic-cell reprogramming. Trends Cell Biol 2013; 24:179-87. [PMID: 24183602 DOI: 10.1016/j.tcb.2013.09.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/20/2013] [Accepted: 09/23/2013] [Indexed: 12/27/2022]
Abstract
Pioneering work over the past years has highlighted the remarkable ability of manipulating cell states through exogenous, mostly transcription factor-induced reprogramming. The use of small molecules and reprogramming by transcription factors share a common history starting with the early AZA and MyoD experiments in fibroblast cells. Recent work shows that a combination of small molecules can replace all of the reprogramming factors and many previous studies have demonstrated their use in enhancing efficiencies or replacing individual factors. Here we provide a brief introduction to reprogramming followed by a detailed review of the major classes of small molecules that have been used to date and what future opportunities can be expected from these.
Collapse
|
34
|
Lin C, Yu C, Ding S. Toward directed reprogramming through exogenous factors. Curr Opin Genet Dev 2013; 23:519-25. [PMID: 23932127 DOI: 10.1016/j.gde.2013.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/10/2013] [Accepted: 06/23/2013] [Indexed: 12/25/2022]
Abstract
Direct reprogramming of one cell type into another provides unprecedented opportunities to study fundamental biology, model disease, and develop regenerative medicine. Different paradigms of reprogramming strategies with different sets of factors have been developed to generate various cell types, including induced pluripotent stem cells, neuronal or neural precursor cells, cardiomyocyte-like cells, endothelial cells, and hepatocyte-like cells. Various exogenous factors, especially small molecules modulating signaling, cellular state, and transcription, have been identified to enhance and enable reprogramming. With an increased understanding of reprogramming mechanisms and discovery of new molecules, it is conceivable that reprogramming can be achieved in a more directed and deterministic manner under entirely chemically defined conditions.
Collapse
Affiliation(s)
- Changsheng Lin
- Gladstone Institute of Cardiovascular Disease, Department of Pharmaceutical Chemistry, University of California, San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | | | | |
Collapse
|
35
|
Abstract
Pluripotent stem cells can differentiate into nearly all types of cells in the body. This unique potential provides significant promise for cell-based therapies to restore tissues or organs destroyed by injuries, degenerative diseases, aging, or cancer. The discovery of induced pluripotent stem cell (iPSC) technology offers a possible strategy to generate patient-specific pluripotent stem cells. However, because of concerns about the specificity, efficiency, kinetics, and safety of iPSC reprogramming, improvements or fundamental changes in this process are required before their effective clinical use. A chemical approach is regarded as a promising strategy to improve and change the iPSC process. Dozens of small molecules have been identified that can functionally replace reprogramming factors and significantly improve iPSC reprogramming. In addition to the prospect of deriving patient-specific tissues and organs from iPSCs, another attractive strategy for regenerative medicine is transdifferentiation-the direct conversion of one somatic cell type to another. Recent studies revealed a new paradigm of transdifferentiation: using transcription factors used in iPSC generation to induce transdifferentiation or called iPSC transcription factor-based transdifferentiation. This type of transdifferentiation not only reveals and uses the developmentally plastic intermediates generated during iPSC reprogramming but also produces a wide range of cells, including expandable tissue-specific precursor cells. Here, we review recent progress of small molecule approaches in the generation of iPSCs. In addition, we summarize the new concept of iPSC transcription factor-based transdifferentiation and discuss its application in generating various lineage-specific cells, especially cardiovascular cells.
Collapse
Affiliation(s)
- Tianhua Ma
- Department of Pharmaceutical Chemistry, Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
36
|
Abstract
The isolation of embryonic stem cells (ESCs) has furthered our understanding of normal embryonic development and fueled the progression of stem cell derived therapies. However, the generation of ESCs requires the destruction of an embryo, making the use of these cells ethically controversial. In 2006 the Yamanaka group overcame this ethical controversy when they described a protocol whereby somatic cells could be dedifferentiated into a pluripotent state following the transduction of a four transcription factor cocktail. Following this initial study numerous groups have described protocols to generate induced pluripotent stem cells (iPSCs). These protocols have simplified the reprogramming strategy by employing polycistronic reprogramming cassettes and flanking such polycistronic cassettes with loxP or piggyBac recognition sequences. Thus, these strategies allow for excision of the entire transgene cassette, limiting the potential for the integration of exogenous transgenes to have detrimental effect. Others have prevented the potentially deleterious effects of integrative reprogramming strategies by using non-integrating adenoviral vectors, traditional recombinant DNA transfection, transfection of minicircle DNA, or transfection of episomally maintained EBNA1/OriP plasmids. Interestingly, transfection of mRNA or miRNA has also been shown to be capable of reprogramming cells, and multiple groups have developed protocols using cell penetrating peptide tagged reprogramming factors to de-differentiate somatic cells in the absence of exogenous nucleic acid. Despite the numerous different reprogramming strategies that have been developed, the reprogramming process remains extremely inefficient. To overcome this inefficiency multiple groups have successfully used small molecules such as valproic acid, sodium butyrate, PD0325901, and others to generate iPSCs.The fast paced field of cellular reprogramming has recently produced protocols to generate iPSCs using non integrative techniques with an ever improving efficiency. These recent developments have brought us one step closer to developing a safe and efficient method to reprogram cells for clinical use. However, a lot of work is still needed before iPSCs can be implemented in a clinical setting.
Collapse
|
37
|
Lee J, Xia Y, Son MY, Jin G, Seol B, Kim MJ, Son MJ, Do M, Lee M, Kim D, Lee K, Cho YS. A Novel Small Molecule Facilitates the Reprogramming of Human Somatic Cells into a Pluripotent State and Supports the Maintenance of an Undifferentiated State of Human Pluripotent Stem Cells. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201206691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
38
|
Lee J, Xia Y, Son MY, Jin G, Seol B, Kim MJ, Son MJ, Do M, Lee M, Kim D, Lee K, Cho YS. A novel small molecule facilitates the reprogramming of human somatic cells into a pluripotent state and supports the maintenance of an undifferentiated state of human pluripotent stem cells. Angew Chem Int Ed Engl 2012; 51:12509-13. [PMID: 23125037 DOI: 10.1002/anie.201206691] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Indexed: 01/11/2023]
Abstract
Booster of pluripotency: RSC133, a new synthetic derivative of indoleacrylic acid/indolepropionic acid, exhibits dual activity by inhibiting histone deacetylase and DNA methyltransferase. Furthermore it potently improves the reprogramming of human somatic cells into a pluripotent state and aids the growth and maintenance of human pluripotent stem cells (hPSCs).
Collapse
Affiliation(s)
- Jungwoon Lee
- Regenerative Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 305-806, S. Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Evans LE, Cheeseman MD, Jones K. N-N bond-forming cyclization for the one-pot synthesis of N-aryl[3,4-d]pyrazolopyrimidines. Org Lett 2012; 14:3546-9. [PMID: 22734502 PMCID: PMC3390909 DOI: 10.1021/ol301561a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
An efficient one-pot synthesis of N-aryl[3,4-d]pyrazolopyrimidines in good yield and under mild reaction conditions is described. By exploiting electron-deficient hydroxylamines, the substituted oxime products were formed with very high E-diastereoselectivity. The key step utilizes a cyclization reaction upon an oxime derived from hydroxylamine-O-sulfonic acid to form the N–N bond of the product.
Collapse
Affiliation(s)
- Lindsay E Evans
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | | |
Collapse
|