1
|
Kohout VR, Wardzala CL, Kramer JR. Synthesis and biomedical applications of mucin mimic materials. Adv Drug Deliv Rev 2022; 191:114540. [PMID: 36228896 PMCID: PMC10066857 DOI: 10.1016/j.addr.2022.114540] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/17/2022] [Accepted: 09/13/2022] [Indexed: 02/09/2023]
Abstract
Mucin glycoproteins are the major component of mucus and coat epithelial cell surfaces forming the glycocalyx. The glycocalyx and mucus are involved in the transport of nutrients, drugs, gases, and pathogens toward the cell surface. Mucins are also involved in diverse diseases such as cystic fibrosis and cancer. Due to inherent heterogeneity in native mucin structure, many synthetic materials have been designed to probe mucin chemistry, biology, and physics. Such materials include various glycopolymers, low molecular weight glycopeptides, glycopolypeptides, polysaccharides, and polysaccharide-protein conjugates. This review highlights advances in the area of design and synthesis of mucin mimic materials, and their biomedical applications in glycan binding, epithelial models of infection, therapeutic delivery, vaccine formulation, and beyond.
Collapse
Affiliation(s)
- Victoria R Kohout
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | - Casia L Wardzala
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA.
| |
Collapse
|
2
|
Quan Y, Wang M, Xu C, Wang X, Wu Y, Qin D, Lin Y, Lu X, Lu F, Li L. Cnot8 eliminates naïve regulation networks and is essential for naïve-to-formative pluripotency transition. Nucleic Acids Res 2022; 50:4414-4435. [PMID: 35390160 PMCID: PMC9071485 DOI: 10.1093/nar/gkac236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 03/11/2022] [Accepted: 03/26/2022] [Indexed: 11/14/2022] Open
Abstract
Mammalian early epiblasts at different phases are characterized by naïve, formative, and primed pluripotency states, involving extensive transcriptome changes. Here, we report that deadenylase Cnot8 of Ccr4-Not complex plays essential roles during the transition from naïve to formative state. Knock out (KO) Cnot8 resulted in early embryonic lethality in mice, but Cnot8 KO embryonic stem cells (ESCs) could be established. Compared with the cells differentiated from normal ESCs, Cnot8 KO cells highly expressed a great many genes during their differentiation into the formative state, including several hundred naïve-like genes enriched in lipid metabolic process and gene expression regulation that may form the naïve regulation networks. Knockdown expression of the selected genes of naïve regulation networks partially rescued the differentiation defects of Cnot8 KO ESCs. Cnot8 depletion led to the deadenylation defects of its targets, increasing their poly(A) tail lengths and half-life, eventually elevating their expression levels. We further found that Cnot8 was involved in the clearance of targets through its deadenylase activity and the binding of Ccr4-Not complex, as well as the interacting with Tob1 and Pabpc1. Our results suggest that Cnot8 eliminates naïve regulation networks through mRNA clearance, and is essential for naïve-to-formative pluripotency transition.
Collapse
Affiliation(s)
- Yujun Quan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meijiao Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100101, China
| | - Chengpeng Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dandan Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuxuan Lin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xukun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Falong Lu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
nr0b1 (DAX1) loss of function in zebrafish causes hypothalamic defects via abnormal progenitor proliferation and differentiation. J Genet Genomics 2021; 49:217-229. [PMID: 34606992 DOI: 10.1016/j.jgg.2021.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 11/23/2022]
Abstract
The nuclear receptor DAX-1 (encoded by the NR0B1 gene) is presented in the hypothalamic tissues in humans and other vertebrates. Human patients with NR0B1 mutations often have hypothalamic-pituitary defects, but the involvement of NR0B1 in hypothalamic development and function is not well understood. Here, we report the disruption of the nr0b1 gene in zebrafish causes abnormal expression of gonadotropins, a reduction in fertilization rate, and an increase in post-fasting food intake, which is indicative of abnormal hypothalamic functions. We find that loss of nr0b1 increases the number of prodynorphin (pdyn)-expressing neurons but decreases the number of pro-opiomelanocortin (pomcb)-expressing neurons in the zebrafish hypothalamic arcuate region (ARC). Further examination reveals that the proliferation of progenitor cells is reduced in the hypothalamus of nr0b1 mutant embryos accompanying with the decreased expression of genes in the Notch signaling pathway. Additionally, the inhibition of Notch signaling in wild-type embryos increases the number of pdyn neurons, mimicking the nr0b1 mutant phenotype. In contrast, ectopic activation of Notch signaling in nr0b1 mutant embryos decreases the number of pdyn neurons. Taken together, our results suggest that nr0b1 regulates neural progenitor proliferation and maintenance to ensure normal hypothalamic neuron development.
Collapse
|
4
|
Hu Z, Li H, Jiang H, Ren Y, Yu X, Qiu J, Stablewski AB, Zhang B, Buck MJ, Feng J. Transient inhibition of mTOR in human pluripotent stem cells enables robust formation of mouse-human chimeric embryos. SCIENCE ADVANCES 2020; 6:eaaz0298. [PMID: 32426495 PMCID: PMC7220352 DOI: 10.1126/sciadv.aaz0298] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 03/02/2020] [Indexed: 06/11/2023]
Abstract
It has not been possible to generate naïve human pluripotent stem cells (hPSCs) that substantially contribute to mouse embryos. We found that a brief inhibition of mTOR with Torin1 converted hPSCs from primed to naïve pluripotency. The naïve hPSCs were maintained in the same condition as mouse embryonic stem cells and exhibited high clonogenicity, rapid proliferation, mitochondrial respiration, X chromosome reactivation, DNA hypomethylation, and transcriptomes sharing similarities to those of human blastocysts. When transferred to mouse blastocysts, naïve hPSCs generated 0.1 to 4% human cells, of all three germ layers, including large amounts of enucleated red blood cells, suggesting a marked acceleration of hPSC development in mouse embryos. Torin1 induced nuclear translocation of TFE3; TFE3 with mutated nuclear localization signal blocked the primed-to-naïve conversion. The generation of chimera-competent naïve hPSCs unifies some common features of naïve pluripotency in mammals and may enable applications such as human organ generation in animals.
Collapse
Affiliation(s)
- Zhixing Hu
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Hanqin Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Houbo Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Yong Ren
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Xinyang Yu
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jingxin Qiu
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Aimee B. Stablewski
- Gene Targeting and Transgenic Shared Resource, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Boyang Zhang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Michael J. Buck
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jian Feng
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
5
|
Eno C, Gomez T, Slusarski DC, Pelegri F. Slow calcium waves mediate furrow microtubule reorganization and germ plasm compaction in the early zebrafish embryo. Development 2018; 145:dev156604. [PMID: 29632136 PMCID: PMC6001370 DOI: 10.1242/dev.156604] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
Zebrafish germ plasm ribonucleoparticles (RNPs) become recruited to furrows of early zebrafish embryos through their association with astral microtubules ends. During the initiation of cytokinesis, microtubules are remodeled into a furrow microtubule array (FMA), which is thought to be analogous to the mammalian midbody involved in membrane abscission. During furrow maturation, RNPs and FMA tubules transition from their original distribution along the furrow to enrichments at the furrow distal ends, which facilitates germ plasm mass compaction. We show that nebel mutants exhibit reduced furrow-associated slow calcium waves (SCWs), caused at least in part by defective enrichment of calcium stores. RNP and FMA distal enrichment mirrors the medial-to-distal polarity of SCWs, and inhibition of calcium release or downstream mediators such as Calmodulin affects RNP and FMA distal enrichment. Blastomeres with reduced or lacking SCWs, such as early blastomeres in nebel mutants and wild-type blastomeres at later stages, exhibit medially bundling microtubules similar to midbodies in other cell types. Our data indicate that SCWs provide medial-to-distal directionality along the furrow to facilitate germ plasm RNP enrichment at the furrow ends.
Collapse
Affiliation(s)
- Celeste Eno
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Timothy Gomez
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Diane C Slusarski
- Department of Biology, The University of Iowa, Iowa City, IA 52242, USA
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, WI 53706, USA
| |
Collapse
|
6
|
Göktuna SI, Diamanti MA, Chau TL. IKK
s and tumor cell plasticity. FEBS J 2018; 285:2161-2181. [DOI: 10.1111/febs.14444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/22/2018] [Accepted: 03/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Serkan I. Göktuna
- Department of Molecular Biology and Genetics Bilkent University Ankara Turkey
- National Nanotechnology Research Center (UNAM) Bilkent University Ankara Turkey
| | - Michaela A. Diamanti
- Georg‐Speyer‐Haus Institute for Tumor Biology and Experimental Therapy Frankfurt am Main Germany
| | - Tieu Lan Chau
- Department of Molecular Biology and Genetics Bilkent University Ankara Turkey
| |
Collapse
|
7
|
Xie X, Fu Y, Liu J. Chemical reprogramming and transdifferentiation. Curr Opin Genet Dev 2017; 46:104-113. [PMID: 28755566 DOI: 10.1016/j.gde.2017.07.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/09/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022]
Abstract
The revolutionizing somatic cell reprogramming/transdifferentiation technologies provide a new path for cell replacement therapies and drug screening. The original method for reprogramming involves the delivery of exogenous transcription factors, leading to the safety concerns about the possible genome integration. Many efforts have been taken to avoid genetic alteration in somatic cell reprogramming/transdifferentiation by using non-integrating gene delivery approaches, cell membrane permeable proteins, and small molecule compounds. Compared to other methods, small-molecule compounds have several unique advantages, such as structural versatility and being easy to control in a time-dependent and concentration-dependent way. More importantly, small molecules have been used as drugs to treat human diseases for thousands of years. So the small molecule approach to reprogramming might be more acceptable in clinical-related uses. In the past few years, small molecule approaches have made significant progresses in inducing pluripotent or functional differentiated cells from somatic cells. Here we review the recent achievements of chemical reprogramming/transdifferentiation and discuss the advantages and challenges facing this strategy in future applications.
Collapse
Affiliation(s)
- Xin Xie
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Yanbin Fu
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jian Liu
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
8
|
Wnt/β-catenin signaling promotes self-renewal and inhibits the primed state transition in naïve human embryonic stem cells. Proc Natl Acad Sci U S A 2016; 113:E6382-E6390. [PMID: 27698112 PMCID: PMC5081574 DOI: 10.1073/pnas.1613849113] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In both mice and humans, pluripotent stem cells (PSCs) exist in at least two distinct states of pluripotency, known as the naïve and primed states. Our understanding of the intrinsic and extrinsic factors that enable PSCs to self-renew and to transition between different pluripotent states is important for understanding early development. In mouse embryonic stem cells (mESCs), Wnt proteins stimulate mESC self-renewal and support the naïve state. In human embryonic stem cells (hESCs), Wnt/β-catenin signaling is active in naïve-state hESCs and is reduced or absent in primed-state hESCs. However, the role of Wnt/β-catenin signaling in naïve hESCs remains largely unknown. Here, we demonstrate that inhibition of the secretion of Wnts or inhibition of the stabilization of β-catenin in naïve hESCs reduces cell proliferation and colony formation. Moreover, we show that addition of recombinant Wnt3a partially rescues cell proliferation in naïve hESCs caused by inhibition of Wnt secretion. Notably, inhibition of Wnt/β-catenin signaling in naïve hESCs did not cause differentiation. Instead, it induced primed hESC-like proteomic and metabolic profiles. Thus, our results suggest that naïve hESCs secrete Wnts that activate autocrine or paracrine Wnt/β-catenin signaling to promote efficient self-renewal and inhibit the transition to the primed state.
Collapse
|
9
|
Mammalian non-CG methylations are conserved and cell-type specific and may have been involved in the evolution of transposon elements. Sci Rep 2016; 6:32207. [PMID: 27573482 PMCID: PMC5004121 DOI: 10.1038/srep32207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/03/2016] [Indexed: 12/04/2022] Open
Abstract
Although non-CG methylations are abundant in several mammalian cell types, their biological significance is sparsely characterized. We gathered 51 human and mouse DNA methylomes from brain neurons, embryonic stem cells and induced pluripotent stem cells, primordial germ cells and oocytes. We utilized an unbiased sub-motif prediction method and reported CW as the representative non-CG methylation context, which is distinct from CC methylation in terms of sequence context and genomic distribution. A two-dimensional comparison of non-CG methylations across cell types and species was performed. Unambiguous studies of sequence preferences and genomic region enrichment showed that CW methylation is cell-type specific and is also conserved between humans and mice. In brain neurons, it was found that active long interspersed nuclear element-1 (LINE-1) lacked CW methylations but not CG methylations. Coincidentally, both human Alu and mouse B1 elements preferred high CW methylations at specific loci during their respective evolutionary development. Last, the strand-specific distributions of CW methylations in introns and long interspersed nuclear elements are also cell-type specific and conserved. In summary, our results illustrate that CW methylations are highly conserved among species, are dynamically regulated in each cell type, and are potentially involved in the evolution of transposon elements.
Collapse
|
10
|
Zhao ZA, Yu Y, Ma HX, Wang XX, Lu X, Zhai Y, Zhang X, Wang H, Li L. The roles of ERAS during cell lineage specification of mouse early embryonic development. Open Biol 2016; 5:rsob.150092. [PMID: 26269429 PMCID: PMC4554925 DOI: 10.1098/rsob.150092] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Eras encodes a Ras-like GTPase protein that was originally identified as an embryonic stem cell-specific Ras. ERAS has been known to be required for the growth of embryonic stem cells and stimulates somatic cell reprogramming, suggesting its roles on mouse early embryonic development. We now report a dynamic expression pattern of Eras during mouse peri-implantation development: its expression increases at the blastocyst stage, and specifically decreases in E7.5 mesoderm. In accordance with its expression pattern, the increased expression of Eras promotes cell proliferation through controlling AKT activation and the commitment from ground to primed state through ERK activation in mouse embryonic stem cells; and the reduced expression of Eras facilitates primitive streak and mesoderm formation through AKT inhibition during gastrulation. The expression of Eras is finely regulated to match its roles in mouse early embryonic development during which Eras expression is negatively regulated by the β-catenin pathway. Thus, beyond its well-known role on cell proliferation, ERAS may also play important roles in cell lineage specification during mouse early embryonic development.
Collapse
Affiliation(s)
- Zhen-Ao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou 215000, People's Republic of China
| | - Yang Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China Institute of Zoology, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huai-Xiao Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China Institute of Zoology, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiao-Xiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China Institute of Zoology, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xukun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China Institute of Zoology, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yanhua Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Xiaoxin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Haibin Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| |
Collapse
|
11
|
Takata N, Sakakura E, Kasukawa T, Sakuma T, Yamamoto T, Sasai Y. Establishment of Functional Genomics Pipeline in Mouse Epiblast-Like Tissue by Combining Transcriptomic Analysis and Gene Knockdown/Knockin/Knockout, Using RNA Interference and CRISPR/Cas9. Hum Gene Ther 2016; 27:436-50. [PMID: 26839115 DOI: 10.1089/hum.2015.148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The epiblast (foremost embryonic ectoderm) generates all three germ layers and therefore has crucial roles in the formation of all mammalian body cells. However, regulation of epiblast gene expression is poorly understood because of the difficulty of manipulating epiblast tissues in vivo. In the present study, using the self-organizing properties of mouse embryonic stem cell (ESC), we generated and characterized epiblast-like tissue in three-dimensional culture. We identified significant genome-wide gene expression changes in this epiblast-like tissue by transcriptomic analysis. In addition, we identified the particular significance of the Erk/Mapk and integrin-linked kinase pathways, and genes related to ectoderm/epithelial formation, using the bioinformatics resources IPA and DAVID. Here, we focused on Fgf5, which ranked in the top 10 among the discovered genes. To develop a functional analysis of Fgf5, we created an efficient method combining CRISPR/Cas9-mediated genome engineering and RNA interference (RNAi). Notably, we show one-step generation of various Fgf5 reporter lines including heterozygous and homozygous knockins (the GET method). For time- and dose-dependent depletion of fgf5 over the course of development, we generated an ESC line harboring Tol2 transposon-mediated integration of an inducible short hairpin RNA interference system (pdiRNAi). Our findings raised the possibility that Fgf/Erk signaling and apicobasal epithelial integrity are important factors in epiblast development. In addition, our methods provide a framework for a broad array of applications in the areas of mammalian genetics and molecular biology to understand development and to improve future therapeutics.
Collapse
Affiliation(s)
- Nozomu Takata
- 1 Laboratory for In Vitro Histogenesis, RIKEN Center for Developmental Biology , Hyogo, Japan
| | - Eriko Sakakura
- 1 Laboratory for In Vitro Histogenesis, RIKEN Center for Developmental Biology , Hyogo, Japan
| | - Takeya Kasukawa
- 2 Large Scale Data Managing Unit, RIKEN Center for Life Science Technologies , Kanagawa, Japan
| | - Tetsushi Sakuma
- 3 Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Takashi Yamamoto
- 3 Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Yoshiki Sasai
- 4 Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology , Hyogo, Japan
| |
Collapse
|
12
|
Van der Jeught M, Taelman J, Duggal G, Ghimire S, Lierman S, Chuva de Sousa Lopes SM, Deforce D, Deroo T, De Sutter P, Heindryckx B. Application Of Small Molecules Favoring Naïve Pluripotency during Human Embryonic Stem Cell Derivation. Cell Reprogram 2016; 17:170-80. [PMID: 26053517 DOI: 10.1089/cell.2014.0085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In mice, inhibition of both the fibroblast growth factor (FGF) mitogen-activated protein kinase kinase/extracellular-signal regulated kinase (MEK/Erk) and the Wnt signaling inhibitor glycogen synthase-3β (GSK3β) enables the derivation of mouse embryonic stem cells (mESCs) from nonpermissive strains in the presence of leukemia inhibitory factor (LIF). Whereas mESCs are in an uncommitted naïve state, human embryonic stem cells (hESCs) represent a more advanced state, denoted as primed pluripotency. This burdens hESCs with a series of characteristics, which, in contrast to naïve ESCs, makes them not ideal for key applications such as cell-based clinical therapies and human disease modeling. In this study, different small molecule combinations were applied during human ESC derivation. Hereby, we aimed to sustain the naïve pluripotent state, by interfering with various key signaling pathways. First, we tested several combinations on existing, 2i (PD0325901 and CHIR99021)-derived mESCs. All combinations were shown to be equally adequate to sustain the expression of naïve pluripotency markers. Second, these conditions were tested during hESC derivation. Overall, the best results were observed in the presence of medium supplemented with 2i, LIF, and the noncanonical Wnt signaling agonist Wnt5A, alone and combined with epinephrine. In these conditions, outgrowths repeatedly showed an ESC progenitor-like morphology, starting from day 3. Culturing these "progenitor cells" did not result in stable, naïve hESC lines in the current conditions. Although Wnt5A could not promote naïve hESC derivation, we found that it was sustaining the conversion of established hESCs toward a more naïve state. Future work should aim to distinct the effects of the various culture formulations, including our Wnt5A-supplemented medium, reported to promote stable naïve pluripotency in hESCs.
Collapse
Affiliation(s)
- Margot Van der Jeught
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium .,4 These authors contributed equally to this work
| | - Jasin Taelman
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium .,4 These authors contributed equally to this work
| | - Galbha Duggal
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| | - Sabitri Ghimire
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| | - Sylvie Lierman
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| | - Susana M Chuva de Sousa Lopes
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium .,2 Department of Anatomy and Embryology, Leiden University Medical Center , 2300 Leiden, The Netherlands
| | - Dieter Deforce
- 3 Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University , 9000 Ghent, Belgium
| | - Tom Deroo
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| | - Petra De Sutter
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| | - Björn Heindryckx
- 1 Ghent Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital , 9000 Ghent, Belgium
| |
Collapse
|
13
|
Canton I, Warren N, Chahal A, Amps K, Wood A, Weightman R, Wang E, Moore H, Armes SP. Mucin-Inspired Thermoresponsive Synthetic Hydrogels Induce Stasis in Human Pluripotent Stem Cells and Human Embryos. ACS CENTRAL SCIENCE 2016; 2:65-74. [PMID: 27163030 PMCID: PMC4827554 DOI: 10.1021/acscentsci.5b00370] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Indexed: 05/07/2023]
Abstract
Human pluripotent stem cells (hPSCs; both embryonic and induced pluripotent) rapidly proliferate in adherent culture to maintain their undifferentiated state. However, for mammals exhibiting delayed gestation (diapause), mucin-coated embryos can remain dormant for days or months in utero, with their constituent PSCs remaining pluripotent under these conditions. Here we report cellular stasis for both hPSC colonies and preimplantation embryos immersed in a wholly synthetic thermoresponsive gel comprising poly(glycerol monomethacrylate)-poly(2-hydroxypropyl methacrylate) [PGMA55-PHPMA135] diblock copolymer worms. This hydroxyl-rich mucin-mimicking nonadherent 3D gel maintained PSC viability and pluripotency in the quiescent G0 state without passaging for at least 14 days. Similarly, gel-coated human embryos remain in a state of suspended animation (diapause) for up to 8 days. The discovery of a cryptic cell arrest mechanism for both hPSCs and embryos suggests an important connection between the cellular mechanisms that evoke embryonic diapause and pluripotency. Moreover, such synthetic worm gels offer considerable utility for the short-term (weeks) storage of either pluripotent stem cells or human embryos without cryopreservation.
Collapse
Affiliation(s)
- Irene Canton
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Nicholas
J. Warren
- Department
of Chemistry, Dainton Building, University
of Sheffield, Sheffield, S3 7HF, U.K.
| | - Aman Chahal
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Katherine Amps
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Andrew Wood
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Richard Weightman
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Eugenia Wang
- Department
of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky 40202, United States
| | - Harry Moore
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, U.K.
| | - Steven P. Armes
- Department
of Chemistry, Dainton Building, University
of Sheffield, Sheffield, S3 7HF, U.K.
| |
Collapse
|
14
|
Jo J, Hwang S, Kim HJ, Hong S, Lee JE, Lee SG, Baek A, Han H, Lee JI, Lee I, Lee DR. An integrated systems biology approach identifies positive cofactor 4 as a factor that increases reprogramming efficiency. Nucleic Acids Res 2016; 44:1203-15. [PMID: 26740582 PMCID: PMC4756831 DOI: 10.1093/nar/gkv1468] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022] Open
Abstract
Spermatogonial stem cells (SSCs) can spontaneously dedifferentiate into embryonic stem cell (ESC)-like cells, which are designated as multipotent SSCs (mSSCs), without ectopic expression of reprogramming factors. Interestingly, SSCs express key pluripotency genes such as Oct4, Sox2, Klf4 and Myc. Therefore, molecular dissection of mSSC reprogramming may provide clues about novel endogenous reprogramming or pluripotency regulatory factors. Our comparative transcriptome analysis of mSSCs and induced pluripotent stem cells (iPSCs) suggests that they have similar pluripotency states but are reprogrammed via different transcriptional pathways. We identified 53 genes as putative pluripotency regulatory factors using an integrated systems biology approach. We demonstrated a selected candidate, Positive cofactor 4 (Pc4), can enhance the efficiency of somatic cell reprogramming by promoting and maintaining transcriptional activity of the key reprograming factors. These results suggest that Pc4 has an important role in inducing spontaneous somatic cell reprogramming via up-regulation of key pluripotency genes.
Collapse
Affiliation(s)
- Junghyun Jo
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea
| | - Sohyun Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | | - Soomin Hong
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea
| | | | - Sung-Geum Lee
- CHA Stem Cell Institute, CHA University, Seoul, Korea
| | - Ahmi Baek
- CHA Stem Cell Institute, CHA University, Seoul, Korea
| | - Heonjong Han
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jin Il Lee
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Dong Ryul Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea CHA Stem Cell Institute, CHA University, Seoul, Korea Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| |
Collapse
|
15
|
Production of human pluripotent stem cell therapeutics under defined xeno-free conditions: progress and challenges. Stem Cell Rev Rep 2015; 11:96-109. [PMID: 25077810 DOI: 10.1007/s12015-014-9544-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent advances on human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) have brought us closer to the realization of their clinical potential. Nonetheless, tissue engineering and regenerative medicine applications will require the generation of hPSC products well beyond the laboratory scale. This also mandates the production of hPSC therapeutics in fully-defined, xeno-free systems and in a reproducible manner. Toward this goal, we summarize current developments in defined media free of animal-derived components for hPSC culture. Bioinspired and synthetic extracellular matrices for the attachment, growth and differentiation of hPSCs are also reviewed. Given that most progress in xeno-free medium and substrate development has been demonstrated in two-dimensional rather than three dimensional culture systems, translation from the former to the latter poses unique difficulties. These challenges are discussed in the context of cultivation platforms of hPSCs as aggregates, on microcarriers or after encapsulation in biocompatible scaffolds.
Collapse
|
16
|
Martínez-Arroyo AM, Míguez-Forján JM, Remohí J, Pellicer A, Medrano JV. Understanding Mammalian Germ Line Development with In Vitro Models. Stem Cells Dev 2015; 24:2101-13. [DOI: 10.1089/scd.2015.0060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Ana M. Martínez-Arroyo
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Jose M. Míguez-Forján
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Jose Remohí
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
| | - Antonio Pellicer
- Fundación Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Jose V. Medrano
- Fundación Instituto Valenciano de Infertilidad (FIVI), Valencia University, INCLIVA, Valencia, Spain
- Fundación Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
17
|
Gokhale PJ, Au-Young JK, Dadi S, Keys DN, Harrison NJ, Jones M, Soneji S, Enver T, Sherlock JK, Andrews PW. Culture adaptation alters transcriptional hierarchies among single human embryonic stem cells reflecting altered patterns of differentiation. PLoS One 2015; 10:e0123467. [PMID: 25875838 PMCID: PMC4397016 DOI: 10.1371/journal.pone.0123467] [Citation(s) in RCA: 18] [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: 07/14/2014] [Accepted: 03/03/2015] [Indexed: 01/17/2023] Open
Abstract
We have used single cell transcriptome analysis to re-examine the substates of early passage, karyotypically Normal, and late passage, karyotypically Abnormal (‘Culture Adapted’) human embryonic stem cells characterized by differential expression of the cell surface marker antigen, SSEA3. The results confirmed that culture adaptation is associated with alterations to the dynamics of the SSEA3(+) and SSEA3(-) substates of these cells, with SSEA3(-) Adapted cells remaining within the stem cell compartment whereas the SSEA3(-) Normal cells appear to have differentiated. However, the single cell data reveal that these substates are characterized by further heterogeneity that changes on culture adaptation. Notably the Adapted population includes cells with a transcriptome substate suggestive of a shift to a more naïve-like phenotype in contrast to the cells of the Normal population. Further, a subset of the Normal SSEA3(+) cells expresses genes typical of endoderm differentiation, despite also expressing the undifferentiated stem cell genes, POU5F1 (OCT4) and NANOG, whereas such apparently lineage-primed cells are absent from the Adapted population. These results suggest that the selective growth advantage gained by genetically variant, culture adapted human embryonic stem cells may derive in part from a changed substate structure that influences their propensity for differentiation.
Collapse
Affiliation(s)
- Paul J. Gokhale
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | - SriVidya Dadi
- ThermoFisher, Foster City, California, United States of America
| | - David N. Keys
- ThermoFisher, Foster City, California, United States of America
| | - Neil J. Harrison
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Mark Jones
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Shamit Soneji
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Tariq Enver
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Jon K. Sherlock
- ThermoFisher, Foster City, California, United States of America
| | - Peter W. Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
Paranjpe SS, Veenstra GJC. Establishing pluripotency in early development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:626-36. [PMID: 25857441 DOI: 10.1016/j.bbagrm.2015.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/26/2015] [Accepted: 03/30/2015] [Indexed: 01/23/2023]
Abstract
The earliest steps of embryonic development involve important changes in chromatin and transcription factor networks, which are orchestrated to establish pluripotent cells that will form the embryo. DNA methylation, histone modifications, the pluripotency regulatory network of transcription factors, maternal factors and newly translated proteins all contribute to these transitions in dynamic ways. Moreover, these dynamics are linked to the onset of zygotic transcription. We will review recent progress in our understanding of chromatin state and regulation of gene expression in the context of embryonic development in vertebrates, in particular mouse, Xenopus and zebrafish. We include work on mouse embryonic stem cells and highlight work that illustrates how early embryonic dynamics establish gene regulatory networks and the state of pluripotency.
Collapse
Affiliation(s)
- Sarita S Paranjpe
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gert Jan C Veenstra
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| |
Collapse
|
19
|
Abstract
Pluripotent cells in embryos are situated near the apex of the hierarchy of developmental potential. They are capable of generating all cell types of the mammalian body proper. Therefore, they are the exemplar of stem cells. In vivo, pluripotent cells exist transiently and become expended within a few days of their establishment. Yet, when explanted into artificial culture conditions, they can be indefinitely propagated in vitro as pluripotent stem cell lines. A host of transcription factors and regulatory genes are now known to underpin the pluripotent state. Nonetheless, how pluripotent cells are equipped with their vast multilineage differentiation potential remains elusive. Consensus holds that pluripotency transcription factors prevent differentiation by inhibiting the expression of differentiation genes. However, this does not explain the developmental potential of pluripotent cells. We have presented another emergent perspective, namely, that pluripotency factors function as lineage specifiers that enable pluripotent cells to differentiate into specific lineages, therefore endowing pluripotent cells with their multilineage potential. Here we provide a comprehensive overview of the developmental biology, transcription factors, and extrinsic signaling associated with pluripotent cells, and their accompanying subtypes, in vitro heterogeneity and chromatin states. Although much has been learned since the appreciation of mammalian pluripotency in the 1950s and the derivation of embryonic stem cell lines in 1981, we will specifically emphasize what currently remains unclear. However, the view that pluripotency factors capacitate differentiation, recently corroborated by experimental evidence, might perhaps address the long-standing question of how pluripotent cells are endowed with their multilineage differentiation potential.
Collapse
Affiliation(s)
- Kyle M. Loh
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| | - Bing Lim
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| | - Lay Teng Ang
- Department of Developmental Biology and the Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Genome Institute of Singapore, Stem Cell & Regenerative Biology Group, Agency for Science, Technology & Research, Singapore; and Department of Medicine and the Beth Israel Deaconess Medical Center, Division of Hematology/Oncology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Dowell KG, Simons AK, Bai H, Kell B, Wang ZZ, Yun K, Hibbs MA. Novel insights into embryonic stem cell self-renewal revealed through comparative human and mouse systems biology networks. Stem Cells 2014; 32:1161-72. [PMID: 24307629 DOI: 10.1002/stem.1612] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/11/2013] [Indexed: 12/25/2022]
Abstract
Embryonic stem cells (ESCs), characterized by their ability to both self-renew and differentiate into multiple cell lineages, are a powerful model for biomedical research and developmental biology. Human and mouse ESCs share many features, yet have distinctive aspects, including fundamental differences in the signaling pathways and cell cycle controls that support self-renewal. Here, we explore the molecular basis of human ESC self-renewal using Bayesian network machine learning to integrate cell-type-specific, high-throughput data for gene function discovery. We integrated high-throughput ESC data from 83 human studies (~1.8 million data points collected under 1,100 conditions) and 62 mouse studies (~2.4 million data points collected under 1,085 conditions) into separate human and mouse predictive networks focused on ESC self-renewal to analyze shared and distinct functional relationships among protein-coding gene orthologs. Computational evaluations show that these networks are highly accurate, literature validation confirms their biological relevance, and reverse transcriptase polymerase chain reaction (RT-PCR) validation supports our predictions. Our results reflect the importance of key regulatory genes known to be strongly associated with self-renewal and pluripotency in both species (e.g., POU5F1, SOX2, and NANOG), identify metabolic differences between species (e.g., threonine metabolism), clarify differences between human and mouse ESC developmental signaling pathways (e.g., leukemia inhibitory factor (LIF)-activated JAK/STAT in mouse; NODAL/ACTIVIN-A-activated fibroblast growth factor in human), and reveal many novel genes and pathways predicted to be functionally associated with self-renewal in each species. These interactive networks are available online at www.StemSight.org for stem cell researchers to develop new hypotheses, discover potential mechanisms involving sparsely annotated genes, and prioritize genes of interest for experimental validation.
Collapse
Affiliation(s)
- Karen G Dowell
- The Jackson Laboratory, Bar Harbor, Maine, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Primate-specific endogenous retrovirus-driven transcription defines naive-like stem cells. Nature 2014; 516:405-9. [PMID: 25317556 DOI: 10.1038/nature13804] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 08/28/2014] [Indexed: 01/21/2023]
Abstract
Naive embryonic stem cells hold great promise for research and therapeutics as they have broad and robust developmental potential. While such cells are readily derived from mouse blastocysts it has not been possible to isolate human equivalents easily, although human naive-like cells have been artificially generated (rather than extracted) by coercion of human primed embryonic stem cells by modifying culture conditions or through transgenic modification. Here we show that a sub-population within cultures of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) manifests key properties of naive state cells. These naive-like cells can be genetically tagged, and are associated with elevated transcription of HERVH, a primate-specific endogenous retrovirus. HERVH elements provide functional binding sites for a combination of naive pluripotency transcription factors, including LBP9, recently recognized as relevant to naivety in mice. LBP9-HERVH drives hESC-specific alternative and chimaeric transcripts, including pluripotency-modulating long non-coding RNAs. Disruption of LBP9, HERVH and HERVH-derived transcripts compromises self-renewal. These observations define HERVH expression as a hallmark of naive-like hESCs, and establish novel primate-specific transcriptional circuitry regulating pluripotency.
Collapse
|
22
|
Sancho-Martinez I, Ocampo A, Izpisua Belmonte JC. Reprogramming by lineage specifiers: blurring the lines between pluripotency and differentiation. Curr Opin Genet Dev 2014; 28:57-63. [PMID: 25461451 DOI: 10.1016/j.gde.2014.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/24/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022]
Abstract
The generation of human induced pluripotent stem cells (iPS) has raised enormous expectations within the biomedical community due to their potential vast implications in regenerative and personalized medicine. However, reprogramming to iPS is still not fully comprehended. Difficulties found in ascribing specific molecular patterns to pluripotent cells (PSCs), and inherent inter-line and intra-line variability between different PSCs need to be resolved. Additionally, and despite multiple assumptions, it remains unclear whether the current in vitro culturing conditions for the maintenance and differentiation of PSCs do indeed recapitulate the developmental processes observed in vivo. As a consequence, basic questions such as what is the actual nature of PSCs remain unanswered and different theories have emerged in regards to the identity of these valuable cell population. Here we discuss on the published theories for defining PSC identity, the implications that the different postulated models have for the reprogramming field as well as speculate on potential future directions that might be opened once a precise knowledge on the nature of PSCs is accomplished.
Collapse
Affiliation(s)
- Ignacio Sancho-Martinez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alejandro Ocampo
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Santin G, Paulis M, Vezzoni P, Pacchiana G, Bottiroli G, Croce AC. Autofluorescence properties of murine embryonic stem cells during spontaneous differentiation phases. Lasers Surg Med 2013; 45:597-607. [PMID: 24114723 DOI: 10.1002/lsm.22182] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVE The autofluorescence (AF) analysis allows in vivo, real-time assessment of cell functional activities, depending on the presence of biomolecules strictly involved in metabolic reactions and acting as endogenous fluorophores. Pluripotent stem cells during differentiation are known to undergo changes in their morphofunctional properties, with particular reference to bioenergetic metabolic signatures involving endogenous fluorophores such as NAD(P)H, flavins, lipofuscin-like lipopigments. Since the development of regenerative therapies based on pluripotent cells requires a careful monitoring of the successful maturation into the desired phenotype, aim of our work is to evaluate the AF potential to assess the differentiation phases in a murine stem cell model. STUDY DESIGN/MATERIALS AND METHODS Mouse embryonic stem cells (ESCs) maintained with and without leukemia inhibitory factor (LIF), embryoid bodies (EBs), and EB-derived cells undergoing spontaneous differentiation toward the hematopoietic lineage have been used as a sample models. Cell AF properties have been characterized upon 366-nm excitation, under living conditions and in the absence of exogenous markers. Imaging, microspectrofluorometric techniques, and spectral fitting analysis based on the spectral parameters of each endogenous fluorophore have been applied to estimate their contribution to the whole cell AF emission spectra. Specific cytochemical labeling has been performed to validate AF data. RESULTS Depending on the differentiation phases, cells undergo changes in morphology, AF distribution patterns, and AF emission spectral profiles. These latter reflect variations in the single endogenous fluorophore contribution to the overall emission. The coenzyme NAD(P)H accounts for up to 80% of the whole spectral area. The free form prevails on the bound one, and their changes have been investigated in terms of NAD(P)Hbound/free and redox ratios. These values vary in agreement with a slow metabolic activity and prevailing glycolytic metabolism in the undifferentiated HM1 cells, an increased metabolic activity still relying on glycolysis during the early differentiation phases, and an increased oxidative phosphorylation in EB and hematopoietic precursor cells. Lipofuscin-like lipopigments decrease following differentiation, and porphyrins contributing for less than 5%, prevail in the more actively differentiating cells. These results reflect the shift between anaerobic and aerobic respiration following differentiation, consistently with a decreased autophagy of cell organelles (i.e., mitochondria, as a strategy reported in the literature to keep the undifferentiated homeostasis state), higher mitochondrial activity with more numerous NADH binding sites and synthesis of heme as prosthetic group of proteins, that is, cytochromes. CONCLUSIONS These data open promising perspectives for the monitoring of stem cells differentiation under living conditions without labeling with exogenous agents (inducing perturbations when used in vivo), or immunomarkers not always available for veterinary and zootechnics, by exploiting endogenous fluorophores as intrinsic biomarkers of cell morphofunctional changes.
Collapse
Affiliation(s)
- Giada Santin
- IGM-CNR and Department of Biology & Biotechnology, University Pavia, Pavia, 27100, Italy
| | | | | | | | | | | |
Collapse
|