Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731934

ABSTRACT

Adult bones are continuously remodeled by the balance between bone resorption by osteoclasts and subsequent bone formation by osteoblasts. Many studies have provided molecular evidence that bone remodeling is under the control of circadian rhythms. Circadian fluctuations have been reported in the serum and urine levels of bone turnover markers, such as digested collagen fragments and bone alkaline phosphatase. Additionally, the expressions of over a quarter of all transcripts in bones show circadian rhythmicity, including the genes encoding master transcription factors for osteoblastogenesis and osteoclastogenesis, osteogenic cytokines, and signaling pathway proteins. Serum levels of calcium, phosphate, parathyroid hormone, and calcitonin also display circadian rhythmicity. Finally, osteoblast- and osteoclast-specific knockout mice targeting the core circadian regulator gene Bmal1 show disrupted bone remodeling, although the results have not always been consistent. Despite these studies, however, establishing a direct link between circadian rhythms and bone remodeling in vivo remains a major challenge. It is nearly impossible to repeatedly collect bone materials from human subjects while following circadian changes. In addition, the differences in circadian gene regulation between diurnal humans and nocturnal mice, the main model organism, remain unclear. Filling the knowledge gap in the circadian regulation of bone remodeling could reveal novel regulatory mechanisms underlying many bone disorders including osteoporosis, genetic diseases, and fracture healing. This is also an important question for the basic understanding of how cell differentiation progresses under the influence of cyclically fluctuating environments.


Subject(s)
Bone Remodeling , Circadian Rhythm , Bone Remodeling/genetics , Animals , Circadian Rhythm/physiology , Circadian Rhythm/genetics , Humans , Osteoblasts/metabolism , Osteogenesis/genetics , Osteoclasts/metabolism , Gene Expression Regulation , Bone and Bones/metabolism
2.
PLoS Biol ; 21(12): e3002419, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048364

ABSTRACT

Circadian regulation of gene expression is prevalent and plays critical roles in cell differentiation. However, its roles in the reprogramming of differentiated cells remain largely unknown. Here, we found that one of the master circadian regulators PER1 promoted virus-mediated reprogramming of mouse embryonic fibroblasts (MEFs) to induced neurons (iNs) and induced pluripotent stem cells (iPSCs). Unexpectedly, PER1 achieved this by repressing inflammatory activation of contaminating macrophages in the MEF culture, rather than by directly modulating the reprogrammability of MEFs. More specifically, we found that transduced viruses activated inflammatory genes in macrophages, such as Tnf encoding TNFα, one of the central inflammatory regulators and an autocrine activator of macrophages. TNFα inhibited iN reprogramming, whereas a TNFα inhibitor promoted iN reprogramming, connecting the inflammatory responses to iN reprogramming. In addition, macrophages were induced to proliferate and mature by non-macrophage cells serving as feeders, which also supported up-regulation of TNFα in macrophages without virus transduction. Furthermore, the 2 inflammatory responses were repressed by the circadian regulator PER1 in macrophages, making reprogrammability dependent on time-of-day of virus transduction. Similar results were obtained with iPSC reprogramming, suggesting a wide occurrence of macrophage-mediated inhibition of cell reprogramming. This study uncovers mechanistic links between cell reprogramming, bystander inflammatory macrophages, and circadian rhythms, which are particularly relevant to in vivo reprogramming and organoid formation incorporating immune cells.


Subject(s)
Induced Pluripotent Stem Cells , Tumor Necrosis Factor-alpha , Animals , Mice , Cell Differentiation , Cellular Reprogramming , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Int J Mol Sci ; 24(15)2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37569682

ABSTRACT

Rheumatoid arthritis (RA) represents one of the best examples of circadian fluctuations in disease severity. Patients with RA experience stiffness, pain, and swelling in afflicted joints in the early morning, which tends to become milder toward the afternoon. This has been primarily explained by the higher blood levels of pro-inflammatory hormones and cytokines, such as melatonin, TNFα, IL-1, and IL-6, in the early morning than in the afternoon as well as insufficient levels of anti-inflammatory cortisol, which rises later in the morning. Clinical importance of the circadian regulation of RA symptoms has been demonstrated by the effectiveness of time-of-day-dependent delivery of therapeutic agents in chronotherapy. The primary inflammatory site in RA is the synovium, where increased macrophages, T cells, and synovial fibroblasts play central roles by secreting pro-inflammatory cytokines, chemokines, and enzymes to stimulate each other, additional immune cells, and osteoclasts, ultimately leading to cartilage and bone erosion. Among these central players, macrophages have been one of the prime targets for the study of the link between circadian rhythms and inflammatory activities. Gene knockout experiments of various core circadian regulators have established that disruption of any core circadian regulators results in hyper- or hypoactivation of inflammatory responses by macrophages when challenged by lipopolysaccharide and bacteria. Although these stimulations are not directly linked to RA etiology, these findings serve as a foundation for further study by providing proof of principle. On the other hand, circadian regulation of osteoclasts, downstream effectors of macrophages, remain under-explored. Nonetheless, circadian expression of the inducers of osteoclastogenesis, such as TNFα, IL-1, and IL-6, as well as the knockout phenotypes of circadian regulators in osteoclasts suggest the significance of the circadian control of osteoclast activity in the pathogenesis of RA. More detailed mechanistic understanding of the circadian regulation of macrophages and osteoclasts in the afflicted joints could add novel local therapeutic options for RA.


Subject(s)
Arthritis, Rheumatoid , Osteoclasts , Humans , Osteoclasts/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Arthritis, Rheumatoid/pathology , Macrophages/metabolism , Cytokines/metabolism , Interleukin-1/metabolism
4.
Methods Cell Biol ; 170: 117-125, 2022.
Article in English | MEDLINE | ID: mdl-35811095

ABSTRACT

Skeletal muscle is a highly regenerative tissue that can efficiently recover from various damages caused by injuries and excessive exercises. In adult muscle, stem cells termed satellite cells are mitotically quiescent but activated upon muscle damages to enter the cell cycle as myogenic precursor cells or myoblasts. After several rounds of cell cycles, they exist the cycle and fuse to each other to form multinucleated myotubes, and eventually mature to become contractile myofibers. Satellite cells can be readily isolated from mouse skeletal muscle with enzymatic digestion and magnetic separation with antibodies against specific surface markers. C2C12 cells are an immortalized mouse myoblast cell line that is commercially available and more readily expandable than primary myoblasts. Both primary myoblasts and C2C12 cells have been extensively used as useful in vitro models for myogenic differentiation. Proper examination of this process requires monitoring specific protein expression in subcellular compartments, which can be accomplished through immunofluorescence staining. This chapter describes the workflow for the isolation of satellite cells from mouse skeletal muscle and subsequent immunofluorescence staining to assess the proliferation and differentiation of primary myoblasts and C2C12 cells.


Subject(s)
Muscle Development , Myoblasts , Animals , Cell Differentiation/physiology , Fluorescent Antibody Technique , Mice , Muscle, Skeletal , Myoblasts/metabolism , Staining and Labeling
5.
J Cell Biol ; 220(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34009269

ABSTRACT

Circadian rhythms regulate cell proliferation and differentiation, but circadian control of tissue regeneration remains elusive at the molecular level. Here, we show that proper myoblast differentiation and muscle regeneration are regulated by the circadian master regulators Per1 and Per2. Depletion of Per1 or Per2 suppressed myoblast differentiation in vitro and muscle regeneration in vivo, demonstrating their nonredundant functions. Both Per1 and Per2 were required for the activation of Igf2, an autocrine promoter of myoblast differentiation, accompanied by Per-dependent recruitment of RNA polymerase II, dynamic histone modifications at the Igf2 promoter and enhancer, and the promoter-enhancer interaction. This circadian epigenetic priming created a preferred time window for initiating myoblast differentiation. Consistently, muscle regeneration was faster if initiated at night, when Per1, Per2, and Igf2 were highly expressed compared with morning. This study reveals the circadian timing as a significant factor for effective muscle cell differentiation and regeneration.


Subject(s)
Circadian Rhythm/genetics , Insulin-Like Growth Factor II/genetics , Period Circadian Proteins/genetics , Regeneration/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Knockout , Muscle, Skeletal/growth & development , Myoblasts/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics
6.
Int J Mol Sci ; 20(9)2019 May 08.
Article in English | MEDLINE | ID: mdl-31071906

ABSTRACT

Circadian rhythms regulate over 40% of protein-coding genes in at least one organ in the body through mechanisms tied to the central circadian clock and to cell-intrinsic auto-regulatory feedback loops. Distinct diurnal differences in regulation of regeneration have been found in several organs, including skin, intestinal, and hematopoietic systems. Each regenerating system contains a complex network of cell types with different circadian mechanisms contributing to regeneration. In this review, we elucidate circadian regeneration mechanisms in the three representative systems. We also suggest circadian regulation of global translational activity as an understudied global regulator of regenerative capacity. A more detailed understanding of the molecular mechanisms underlying circadian regulation of tissue regeneration would accelerate the development of new regenerative therapies.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Regeneration/genetics , Ribosomes/genetics , Animals , Hematopoietic System/growth & development , Humans , Intestines/growth & development , Skin/growth & development
7.
Cell Rep ; 25(9): 2537-2551.e8, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30485818

ABSTRACT

Fkbp5 is a widely expressed peptidyl prolyl isomerase that serves as a molecular chaperone through conformational changes of binding partners. Although it regulates diverse protein functions, little is known about its roles in myogenesis. We found here that Fkbp5 plays critical roles in myoblast differentiation through two mechanisms. First, it sequesters Cdk4 within the Hsp90 storage complex and prevents the formation of the cyclin D1-Cdk4 complex, which is a major inhibitor of differentiation. Second, Fkbp5 promotes cis-trans isomerization of the Thr172-Pro173 peptide bond in Cdk4 and inhibits phosphorylation of Thr172, an essential step for Cdk4 activation. Consistent with these in vitro findings, muscle regeneration is delayed in Fkbp5-/- mice. The related protein Fkbp4 also sequesters Cdk4 within the Hsp90 complex but does not isomerize Cdk4 or induce Thr173 phosphorylation despite its highly similar sequence. This study demonstrates protein isomerization as a critical regulatory mechanism of myogenesis by targeting Cdk4.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 4/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Cell Line , Cell Proliferation , HSP90 Heat-Shock Proteins/metabolism , Isomerism , Male , Mice, Knockout , Muscles/physiology , Peptides/metabolism , Proline/metabolism , Protein Binding , Regeneration , Tacrolimus Binding Proteins/deficiency
8.
Cell Rep ; 22(8): 2118-2132, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466738

ABSTRACT

Circadian rhythms regulate cell proliferation and differentiation; however, little is known about their roles in myogenic differentiation. Our synchronized differentiation studies demonstrate that myoblast proliferation and subsequent myotube formation by cell fusion occur in circadian manners. We found that one of the core regulators of circadian rhythms, Cry2, but not Cry1, is critical for the circadian patterns of these two critical steps in myogenic differentiation. This is achieved through the specific interaction between Cry2 and Bclaf1, which stabilizes mRNAs encoding cyclin D1, a G1/S phase transition regulator, and Tmem176b, a transmembrane regulator for myogenic cell fusion. Myoblasts lacking Cry2 display premature cell cycle exit and form short myotubes because of inefficient cell fusion. Consistently, muscle regeneration is impaired in Cry2-/- mice. Bclaf1 knockdown recapitulated the phenotypes of Cry2 knockdown: early cell cycle exit and inefficient cell fusion. This study uncovers a post-transcriptional regulation of myogenic differentiation by circadian rhythms.


Subject(s)
Cell Differentiation , Circadian Rhythm , Cryptochromes/metabolism , Cyclin D1/genetics , Membrane Proteins/metabolism , Muscle Development , RNA Stability/genetics , Repressor Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Fusion , Cell Line , Cyclin D1/metabolism , Gene Expression Regulation , Mice, Knockout , Muscles/metabolism , Myoblasts/cytology , Myoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regeneration
9.
J Stem Cell Res Ther ; 6(3)2016 Mar.
Article in English | MEDLINE | ID: mdl-27158559

ABSTRACT

RNA polymerase II (Pol II) temporarily stops transcription after synthesizing 30-50 bases, and resumes elongation only after stimulations by various signaling molecules and developmental cues. This phenomenon, called promoter-proximal pausing, is observed in 10-50% of the entire genes from Drosophila embryos to human cells. Release of paused Pol II is primarily mediated by the activated form of positive transcription elongation factor b (P-TEFb) initially sequestered in the inhibitory 7SK small nuclear ribonucleoprotein (7SK snRNP) complex. Many proteins and RNAs have been discovered and studied in detail to explain the process of the pausing and release of Pol II in relation to P-TEFb. At the functional level, promoter-proximal pausing regulates genes involved in stimulus-response and development in Drosophila. In mammalian stem cell biology, pausing is important for proliferation and signaling in embryonic stem cells and the formation of induced pluripotent stem cells. Other than this, however, little is known about the biological significance of pausing in mammalian cell differentiation. Further study on pausing mechanisms as well as its functions will contribute to the development of stem cell biology and its clinical applications.

10.
Article in English | MEDLINE | ID: mdl-27158681

ABSTRACT

Faithful duplication of a cell's epigenetic state during DNA replication is essential for the maintenance of a cell's lineage. One of the key mechanisms is the recruitment of several critical chromatin modifying enzymes to the replication fork by proliferating cell nuclear antigen (PCNA). Another mechanism is mediated by the dual function of some histone modifying enzymes as both "reader" and "writer" of the same modification. This capacity allows for parental histones to act as a seed to copy the modification onto nearby newly synthesized histones. In contrast to the vast quantity of research into the maintenance of epigenetic memory, little is known about how the recruitment of these maintenance enzymes changes during stem cell differentiation. This question is especially pertinent due to the recent emphasis on cell reprogramming for regenerative medicine.

11.
Genom Data ; 7: 171-2, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26981399

ABSTRACT

Genome-wide expression patterns of mRNA were compared between mouse embryonic fibroblasts (MEFs), embryonic stem cells (ESCs), and various types of induced pluripotent stem cells (iPSCs). iPSCs were established and maintained using modified Oct4 with or without exogenous leukemia inhibitory factor (LIF) and used to identify mRNAs that were potentially involved in the LIF-independence. The data have been deposited in the NCBI's Gene Expression Omnibus (GEO) database with the accession number GSE65563.

12.
Stem Cell Res ; 15(3): 469-480, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26413786

ABSTRACT

Leukemia inhibitory factor (LIF) is widely used to establish and maintain naïve pluripotent stem cells, including mouse embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Although the combination of chemical inhibitors called 2i can establish mouse iPSCs without LIF from primed pluripotent stem cells, it has been difficult, if not impossible, to establish mouse iPSCs from differentiated somatic cells without LIF. We previously showed that the fusion gene of the transactivation domain of MyoD and the full-length Oct4 (M3O) increases the efficiency of making iPSCs when transduced into fibroblasts along with Sox2, Klf4, and c-Myc (M3O-SKM). Here, we report that M3O-SKM allows for establishment of iPSCs without exogenous LIF from mouse embryonic fibroblasts. The established iPSCs remained undifferentiated and maintained pluripotency over 90 days without LIF as long as M3O was expressed. The iPSCs upregulated miR-205-5p, which was potentially involved in the LIF-independence by suppressing the two signaling pathways inhibited by 2i. The result indicates that potentiated Oct4 can substitute for the LIF signaling pathway, providing a novel model to link Oct4 and LIF, two of the most significant players in naïve pluripotency.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leukemia Inhibitory Factor/pharmacology , MicroRNAs/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Animals , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mice , Mice, Transgenic , Signal Transduction , Transfection
13.
Stem Cell Res ; 15(2): 384-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26318720

ABSTRACT

It has been very difficult, if not impossible, to establish mouse induced pluripotent stem cells (iPSCs) from differentiated cells, such as fibroblasts, without leukemia inhibitory factor (LIF). We have established and maintained LIF-independent iPSCs for longer than 120 days with modified Oct4 along with Sox2, Klf4, and c-Myc. The iPSCs will provide a novel tool to investigate the roles of the LIF-Stat3 signaling pathway in mouse pluripotent stem cells.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leukemia Inhibitory Factor/metabolism , Octamer Transcription Factor-3/metabolism , Animals , Cell Line , Cellular Reprogramming , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , Karyotyping , Kinesins/genetics , Kinesins/metabolism , Kruppel-Like Factor 4 , Leukemia Inhibitory Factor/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
14.
Transl Res ; 165(1): 18-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24695097

ABSTRACT

The recent progress in pluripotent stem cell research has opened new avenues of disease modeling, drug screening, and transplantation of patient-specific tissues unimaginable until a decade ago. The central mechanism underlying pluripotency is epigenetic gene regulation; the majority of cell signaling pathways, both extracellular and cytoplasmic, alter, eventually, the epigenetic status of their target genes during the process of activating or suppressing the genes to acquire or maintain pluripotency. It has long been thought that the chromatin of pluripotent stem cells is open globally to enable the timely activation of essentially all genes in the genome during differentiation into multiple lineages. The current article reviews descriptive observations and the epigenetic machinery relevant to what is supposed to be globally open chromatin in pluripotent stem cells, including microscopic appearance, permissive gene transcription, chromatin remodeling complexes, histone modifications, DNA methylation, noncoding RNAs, dynamic movement of chromatin proteins, nucleosome accessibility and positioning, and long-range chromosomal interactions. Detailed analyses of each element, however, have revealed that the globally open chromatin hypothesis is not necessarily supported by some of the critical experimental evidence, such as genomewide nucleosome accessibility and nucleosome positioning. Greater understanding of epigenetic gene regulation is expected to determine the true nature of the so-called globally open chromatin in pluripotent stem cells.


Subject(s)
Chromatin/genetics , Epigenesis, Genetic , Pluripotent Stem Cells/metabolism , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , Histones/metabolism , Humans , Mice , Nucleosomes/metabolism , RNA, Untranslated/genetics , Transcription, Genetic , Translational Research, Biomedical
16.
Transl Res ; 163(4): 286-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24231336

ABSTRACT

The use of stem cells in the treatment of various diseases and injuries has received increasing interest during the past decade. Injected stem cells, such as mesenchymal stem cells, stimulate tissue repair largely through the secretion of soluble factors that regulate various processes of tissue regeneration, including inflammatory responses, apoptosis, host cell proliferation, and angiogenesis. Recently, it has become apparent that stem cells also use membranous small vesicles, collectively called microvesicles, to repair damaged tissues. Microvesicles are released by many types of cells and exist in almost all types of body fluids. They serve as a vehicle to transfer protein, messenger RNA, and micro RNA to distant cells, altering the gene expression, proliferation, and differentiation of the recipient cells. Although animal models and in vitro studies have suggested promising applications for microvesicles-based regeneration therapy, its effectiveness and feasibility in clinical medicine remain to be established. Further studies of the basic mechanisms responsible for microvesicle-mediated tissue regeneration could lead to novel approaches in regenerative medicine.


Subject(s)
Stem Cell Transplantation/trends , Tissue Engineering/trends , Animals , Humans , Regeneration/physiology , Regenerative Medicine/trends
17.
Cardiovasc Res ; 100(1): 105-13, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23794713

ABSTRACT

AIMS: Fibroblasts can be directly reprogrammed to cardiomyocyte-like cells by introducing defined genes. However, the reprogramming efficiency remains low, delaying the clinical application of this strategy to regenerative cardiology. We previously showed that fusion of the MyoD transactivation domain to the pluripotency transcription factor Oct4 facilitated the transcriptional activity of Oct4, resulting in highly efficient production of induced pluripotent stem cells. We examined whether the same approach can be applied to cardiac transcription factors to facilitate cardiac reprogramming. METHODS AND RESULTS: We fused the MyoD domain to Mef2c, Gata4, Hand2, and Tbx5 and transduced these genes in various combinations into mouse non-cardiac fibroblasts. Transduction of the chimeric Mef2c with the wild-types of the other three genes produced much larger beating clusters of cardiomyocyte-like cells faster than the combination of the four wild-type genes, with an efficiency of 3.5%, >15-fold greater than the wild-type genes. CONCLUSION: Fusion of a powerful transactivation domain to heterologous factors can increase the efficiency of direct reprogramming of fibroblasts to cardiomyocytes.


Subject(s)
Cell Differentiation , Fibroblasts/cytology , MyoD Protein/physiology , Myocytes, Cardiac/cytology , Transcriptional Activation , Animals , Fluorescent Antibody Technique , Induced Pluripotent Stem Cells , MEF2 Transcription Factors/physiology , Mice , MyoD Protein/chemistry , Octamer Transcription Factor-3/physiology , Protein Structure, Tertiary
18.
Cell Biosci ; 2(1): 37, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23126680

ABSTRACT

Long noncoding RNAs (lncRNAs) have been detected in nearly every cell type and found to be fundamentally involved in many biological processes. The characterization of lncRNAs has immense potential to advance our comprehensive understanding of cellular processes and gene regulation, along with implications for the treatment of human disease. The recent ENCODE (Encyclopedia of DNA Elements) study reported 9,640 lncRNA loci in the human genome, which corresponds to around half the number of protein-coding genes. Because of this sheer number and their functional diversity, it is crucial to identify a pool of potentially relevant lncRNAs early on in a given study. In this review, we evaluate the methods for isolating lncRNAs by immunoprecipitation and review the advantages, disadvantages, and applications of three widely used approaches - microarray, tiling array, and RNA-seq - for identifying lncRNAs involved in gene regulation. We also look at ways in which data from publicly available databases such as ENCODE can support the study of lncRNAs.

19.
PLoS One ; 7(6): e39022, 2012.
Article in English | MEDLINE | ID: mdl-22720020

ABSTRACT

Human induced pluripotent stem cells (iPSCs) can be divided into a leukemia inhibitory factor (LIF)-dependent naïve type and a basic fibroblast growth factor (bFGF)-dependent primed type. Although the former are more undifferentiated than the latter, they require signal transduction inhibitors and sustained expression of the transgenes used for iPSC production. We used a transcriptionally enhanced version of OCT4 to establish LIF-dependent human iPSCs without the use of inhibitors and sustained transgene expression. These cells belong to the primed type of pluripotent stem cell, similar to bFGF-dependent iPSCs. Thus, the particular cytokine required for iPSC production does not necessarily define stem cell phenotypes as previously thought. It is likely that the bFGF and LIF signaling pathways converge on unidentified OCT4 target genes. These findings suggest that our LIF-dependent human iPSCs could provide a novel model to investigate the role of cytokine signaling in cellular reprogramming.


Subject(s)
Fibroblast Growth Factors/physiology , Leukemia Inhibitory Factor/physiology , Pluripotent Stem Cells/cytology , Cells, Cultured , Humans , Karyotyping , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
20.
PLoS One ; 7(3): e34149, 2012.
Article in English | MEDLINE | ID: mdl-22479546

ABSTRACT

A major difficulty of producing induced pluripotent stem cells (iPSCs) has been the low efficiency of reprogramming differentiated cells into pluripotent cells. We previously showed that 5% of mouse embryonic fibroblasts (MEFs) were reprogrammed into iPSCs when they were transduced with a fusion gene composed of Oct4 and the transactivation domain of MyoD (called M(3)O), along with Sox2, Klf4 and c-Myc (SKM). In addition, M(3)O facilitated chromatin remodeling of pluripotency genes in the majority of transduced MEFs, including cells that did not become iPSCs. These observations suggested the possibility that more than 5% of cells had acquired the ability to become iPSCs given more favorable culture conditions. Here, we raised the efficiency of making mouse iPSCs with M(3)O-SKM to 26% by culturing transduced cells at low density in serum-free culture medium. In contrast, the efficiency increased from 0.1% to only 2% with the combination of wild-type Oct4 and SKM (OSKM) under the same culture condition. For human iPSCs, M(3)O-SKM achieved 7% efficiency under a similar serum-free culture condition, in comparison to 1% efficiency with OSKM. This study highlights the power of combining the transactivation domain of MyoD with a favorable culture environment.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , MyoD Protein/metabolism , Alkaline Phosphatase/metabolism , Animals , Chromatin Assembly and Disassembly , Coculture Techniques/methods , Culture Media, Serum-Free/pharmacology , Fibroblasts/cytology , Humans , Kruppel-Like Factor 4 , Mice , Mice, Transgenic , Microscopy, Fluorescence/methods , Myoblasts/cytology , Octamer Transcription Factor-3/metabolism , Protein Structure, Tertiary , Teratoma/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...