Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell ; 173(1): 104-116.e12, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29502971

ABSTRACT

Human diseases are often caused by loss of somatic cells that are incapable of re-entering the cell cycle for regenerative repair. Here, we report a combination of cell-cycle regulators that induce stable cytokinesis in adult post-mitotic cells. We screened cell-cycle regulators expressed in proliferating fetal cardiomyocytes and found that overexpression of cyclin-dependent kinase 1 (CDK1), CDK4, cyclin B1, and cyclin D1 efficiently induced cell division in post-mitotic mouse, rat, and human cardiomyocytes. Overexpression of the cell-cycle regulators was self-limiting through proteasome-mediated degradation of the protein products. In vivo lineage tracing revealed that 15%-20% of adult cardiomyocytes expressing the four factors underwent stable cell division, with significant improvement in cardiac function after acute or subacute myocardial infarction. Chemical inhibition of Tgf-ß and Wee1 made CDK1 and cyclin B dispensable. These findings reveal a discrete combination of genes that can efficiently unlock the proliferative potential in cells that have terminally exited the cell cycle.


Subject(s)
Heart/physiology , Myocytes, Cardiac/metabolism , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Proliferation , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cytokinesis , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/veterinary , Myocytes, Cardiac/cytology , Myosin Heavy Chains/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Regeneration , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
2.
Cell ; 167(7): 1734-1749.e22, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984724

ABSTRACT

Mutation of highly conserved residues in transcription factors may affect protein-protein or protein-DNA interactions, leading to gene network dysregulation and human disease. Human mutations in GATA4, a cardiogenic transcription factor, cause cardiac septal defects and cardiomyopathy. Here, iPS-derived cardiomyocytes from subjects with a heterozygous GATA4-G296S missense mutation showed impaired contractility, calcium handling, and metabolic activity. In human cardiomyocytes, GATA4 broadly co-occupied cardiac enhancers with TBX5, another transcription factor that causes septal defects when mutated. The GATA4-G296S mutation disrupted TBX5 recruitment, particularly to cardiac super-enhancers, concomitant with dysregulation of genes related to the phenotypic abnormalities, including cardiac septation. Conversely, the GATA4-G296S mutation led to failure of GATA4 and TBX5-mediated repression at non-cardiac genes and enhanced open chromatin states at endothelial/endocardial promoters. These results reveal how disease-causing missense mutations can disrupt transcriptional cooperativity, leading to aberrant chromatin states and cellular dysfunction, including those related to morphogenetic defects.


Subject(s)
GATA4 Transcription Factor/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Chromatin , Enhancer Elements, Genetic , Female , Heart/growth & development , Humans , Induced Pluripotent Stem Cells , Male , Mutation, Missense , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , T-Box Domain Proteins/genetics
3.
Cell ; 145(2): 183-97, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21477851

ABSTRACT

The embryonic stem (ES) cell transcriptional and chromatin-modifying networks are critical for self-renewal maintenance. However, it remains unclear whether these networks functionally interact and, if so, what factors mediate such interactions. Here, we show that WD repeat domain 5 (Wdr5), a core member of the mammalian Trithorax (trxG) complex, positively correlates with the undifferentiated state and is a regulator of ES cell self-renewal. We demonstrate that Wdr5, an "effector" of H3K4 methylation, interacts with the pluripotency transcription factor Oct4. Genome-wide protein localization and transcriptome analyses demonstrate overlapping gene regulatory functions between Oct4 and Wdr5. The Oct4-Sox2-Nanog circuitry and trxG cooperate in activating transcription of key self-renewal regulators, and furthermore, Wdr5 expression is required for the efficient formation of induced pluripotent stem (iPS) cells. We propose an integrated model of transcriptional and epigenetic control, mediated by select trxG members, for the maintenance of ES cell self-renewal and somatic cell reprogramming.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Proteins/metabolism , Animals , Chromatin Immunoprecipitation , Embryonic Stem Cells/cytology , Histone-Lysine N-Methyltransferase , Histones/metabolism , Intracellular Signaling Peptides and Proteins , Methylation , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Octamer Transcription Factor-3/metabolism , Sequence Analysis, DNA , Transcriptional Activation
4.
Circulation ; 146(10): 770-787, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35938400

ABSTRACT

BACKGROUND: GATA4 (GATA-binding protein 4), a zinc finger-containing, DNA-binding transcription factor, is essential for normal cardiac development and homeostasis in mice and humans, and mutations in this gene have been reported in human heart defects. Defects in alternative splicing are associated with many heart diseases, yet relatively little is known about how cell type- or cell state-specific alternative splicing is achieved in the heart. Here, we show that GATA4 regulates cell type-specific splicing through direct interaction with RNA and the spliceosome in human induced pluripotent stem cell-derived cardiac progenitors. METHODS: We leveraged a combination of unbiased approaches including affinity purification of GATA4 and mass spectrometry, enhanced cross-linking with immunoprecipitation, electrophoretic mobility shift assays, in vitro splicing assays, and unbiased transcriptomic analysis to uncover GATA4's novel function as a splicing regulator in human induced pluripotent stem cell-derived cardiac progenitors. RESULTS: We found that GATA4 interacts with many members of the spliceosome complex in human induced pluripotent stem cell-derived cardiac progenitors. Enhanced cross-linking with immunoprecipitation demonstrated that GATA4 also directly binds to a large number of mRNAs through defined RNA motifs in a sequence-specific manner. In vitro splicing assays indicated that GATA4 regulates alternative splicing through direct RNA binding, resulting in functionally distinct protein products. Correspondingly, knockdown of GATA4 in human induced pluripotent stem cell-derived cardiac progenitors resulted in differential alternative splicing of genes involved in cytoskeleton organization and calcium ion import, with functional consequences associated with the protein isoforms. CONCLUSIONS: This study shows that in addition to its well described transcriptional function, GATA4 interacts with members of the spliceosome complex and regulates cell type-specific alternative splicing via sequence-specific interactions with RNA. Several genes that have splicing regulated by GATA4 have functional consequences and many are associated with dilated cardiomyopathy, suggesting a novel role for GATA4 in achieving the necessary cardiac proteome in normal and stress-responsive conditions.


Subject(s)
GATA4 Transcription Factor , Induced Pluripotent Stem Cells , Alternative Splicing , Animals , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , RNA/genetics , RNA/metabolism
5.
Circ Res ; 127(1): 184-201, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32717173

ABSTRACT

Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia and cause of significant morbidity and mortality. Its increasing prevalence in aging societies constitutes a growing challenge to global healthcare systems. Despite substantial unmet needs in AF prevention and treatment, drug developments hitherto have been challenging, and the current pharmaceutical pipeline is nearly empty. In this review, we argue that current drugs for AF are inadequate because of an oversimplified system for patient classification and the development of drugs that do not interdict underlying disease mechanisms. We posit that an improved understanding of AF molecular pathophysiology related to the continuous identification of novel disease-modifying drug targets and an increased appreciation of patient heterogeneity provide a new framework to personalize AF drug development. Together with recent innovations in diagnostics, remote rhythm monitoring, and big data capabilities, we anticipate that adoption of a new framework for patient subsegmentation based on pathophysiological, genetic, and molecular subsets will improve success rates of clinical trials and advance drugs that reduce the individual patient and public health burden of AF.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Drug Development/methods , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Humans , Molecular Targeted Therapy/methods
6.
Circ Res ; 120(10): 1572-1583, 2017 May 12.
Article in English | MEDLINE | ID: mdl-28400398

ABSTRACT

RATIONALE: During each beat, cardiac myocytes (CMs) generate the mechanical output necessary for heart function through contractile mechanisms that involve shortening of sarcomeres along myofibrils. Human-induced pluripotent stem cells (hiPSCs) can be differentiated into CMs (hiPSC-CMs) that model cardiac contractile mechanical output more robustly when micropatterned into physiological shapes. Quantifying the mechanical output of these cells enables us to assay cardiac activity in a dish. OBJECTIVE: We sought to develop a computational platform that integrates analytic approaches to quantify the mechanical output of single micropatterned hiPSC-CMs from microscopy videos. METHODS AND RESULTS: We micropatterned single hiPSC-CMs on deformable polyacrylamide substrates containing fluorescent microbeads. We acquired videos of single beating cells, of microbead displacement during contractions, and of fluorescently labeled myofibrils. These videos were independently analyzed to obtain parameters that capture the mechanical output of the imaged single cells. We also developed novel methods to quantify sarcomere length from videos of moving myofibrils and to analyze loss of synchronicity of beating in cells with contractile defects. We tested this computational platform by detecting variations in mechanical output induced by drugs and in cells expressing low levels of myosin-binding protein C. CONCLUSIONS: Our method can measure the cardiac function of single micropatterned hiPSC-CMs and determine contractile parameters that can be used to elucidate mechanisms that underlie variations in CM function. This platform will be amenable to future studies of the effects of mutations and drugs on cardiac function.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Multimodal Imaging/methods , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Cells, Cultured , Humans
7.
Circulation ; 135(10): 978-995, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-27834668

ABSTRACT

BACKGROUND: Reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells in situ represents a promising strategy for cardiac regeneration. A combination of 3 cardiac transcription factors, Gata4, Mef2c, and Tbx5 (GMT), can convert fibroblasts into induced cardiomyocyte-like cells, albeit with low efficiency in vitro. METHODS: We screened 5500 compounds in primary cardiac fibroblasts to identify the pathways that can be modulated to enhance cardiomyocyte reprogramming. RESULTS: We found that a combination of the transforming growth factor-ß inhibitor SB431542 and the WNT inhibitor XAV939 increased reprogramming efficiency 8-fold when added to GMT-overexpressing cardiac fibroblasts. The small molecules also enhanced the speed and quality of cell conversion; we observed beating cells as early as 1 week after reprogramming compared with 6 to 8 weeks with GMT alone. In vivo, mice exposed to GMT, SB431542, and XAV939 for 2 weeks after myocardial infarction showed significantly improved reprogramming and cardiac function compared with those exposed to only GMT. Human cardiac reprogramming was similarly enhanced on transforming growth factor-ß and WNT inhibition and was achieved most efficiently with GMT plus myocardin. CONCLUSIONS: Transforming growth factor-ß and WNT inhibitors jointly enhance GMT-induced direct cardiac reprogramming from cardiac fibroblasts in vitro and in vivo and provide a more robust platform for cardiac regeneration.


Subject(s)
Benzamides/pharmacology , Cellular Reprogramming/drug effects , Dioxoles/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Transcription Factors/metabolism , Animals , Benzamides/therapeutic use , Cells, Cultured , Dioxoles/therapeutic use , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart/diagnostic imaging , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Magnetic Resonance Imaging , Mice , Myocardial Infarction/drug therapy , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 112(41): 12705-10, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417073

ABSTRACT

Single cardiomyocytes contain myofibrils that harbor the sarcomere-based contractile machinery of the myocardium. Cardiomyocytes differentiated from human pluripotent stem cells (hPSC-CMs) have potential as an in vitro model of heart activity. However, their fetal-like misalignment of myofibrils limits their usefulness for modeling contractile activity. We analyzed the effects of cell shape and substrate stiffness on the shortening and movement of labeled sarcomeres and the translation of sarcomere activity to mechanical output (contractility) in live engineered hPSC-CMs. Single hPSC-CMs were cultured on polyacrylamide substrates of physiological stiffness (10 kPa), and Matrigel micropatterns were used to generate physiological shapes (2,000-µm(2) rectangles with length:width aspect ratios of 5:1-7:1) and a mature alignment of myofibrils. Translation of sarcomere shortening to mechanical output was highest in 7:1 hPSC-CMs. Increased substrate stiffness and applied overstretch induced myofibril defects in 7:1 hPSC-CMs and decreased mechanical output. Inhibitors of nonmuscle myosin activity repressed the assembly of myofibrils, showing that subcellular tension drives the improved contractile activity in these engineered hPSC-CMs. Other factors associated with improved contractility were axially directed calcium flow, systematic mitochondrial distribution, more mature electrophysiology, and evidence of transverse-tubule formation. These findings support the potential of these engineered hPSC-CMs as powerful models for studying myocardial contractility at the cellular level.


Subject(s)
Cell Differentiation , Cell Shape , Models, Biological , Myocardial Contraction , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Calcium Signaling , Cells, Cultured , Humans , Mitochondria, Heart , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology
9.
Nature ; 465(7299): 808-12, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20535210

ABSTRACT

The generation of reprogrammed induced pluripotent stem cells (iPSCs) from patients with defined genetic disorders holds the promise of increased understanding of the aetiologies of complex diseases and may also facilitate the development of novel therapeutic interventions. We have generated iPSCs from patients with LEOPARD syndrome (an acronym formed from its main features; that is, lentigines, electrocardiographic abnormalities, ocular hypertelorism, pulmonary valve stenosis, abnormal genitalia, retardation of growth and deafness), an autosomal-dominant developmental disorder belonging to a relatively prevalent class of inherited RAS-mitogen-activated protein kinase signalling diseases, which also includes Noonan syndrome, with pleomorphic effects on several tissues and organ systems. The patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase. The iPSCs have been extensively characterized and produce multiple differentiated cell lineages. A major disease phenotype in patients with LEOPARD syndrome is hypertrophic cardiomyopathy. We show that in vitro-derived cardiomyocytes from LEOPARD syndrome iPSCs are larger, have a higher degree of sarcomeric organization and preferential localization of NFATC4 in the nucleus when compared with cardiomyocytes derived from human embryonic stem cells or wild-type iPSCs derived from a healthy brother of one of the LEOPARD syndrome patients. These features correlate with a potential hypertrophic state. We also provide molecular insights into signalling pathways that may promote the disease phenotype.


Subject(s)
Induced Pluripotent Stem Cells/pathology , LEOPARD Syndrome/pathology , Models, Biological , Precision Medicine , Adult , Cell Differentiation , Cell Line , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Enzyme Activation , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/metabolism , LEOPARD Syndrome/drug therapy , LEOPARD Syndrome/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Phosphoproteins/analysis , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , SOXB1 Transcription Factors/genetics
10.
PLoS Comput Biol ; 10(8): e1003777, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25122140

ABSTRACT

A 30-node signed and directed network responsible for self-renewal and pluripotency of mouse embryonic stem cells (mESCs) was extracted from several ChIP-Seq and knockdown followed by expression prior studies. The underlying regulatory logic among network components was then learned using the initial network topology and single cell gene expression measurements from mESCs cultured in serum/LIF or serum-free 2i/LIF conditions. Comparing the learned network regulatory logic derived from cells cultured in serum/LIF vs. 2i/LIF revealed differential roles for Nanog, Oct4/Pou5f1, Sox2, Esrrb and Tcf3. Overall, gene expression in the serum/LIF condition was more variable than in the 2i/LIF but mostly consistent across the two conditions. Expression levels for most genes in single cells were bimodal across the entire population and this motivated a Boolean modeling approach. In silico predictions derived from removal of nodes from the Boolean dynamical model were validated with experimental single and combinatorial RNA interference (RNAi) knockdowns of selected network components. Quantitative post-RNAi expression level measurements of remaining network components showed good agreement with the in silico predictions. Computational removal of nodes from the Boolean network model was also used to predict lineage specification outcomes. In summary, data integration, modeling, and targeted experiments were used to improve our understanding of the regulatory topology that controls mESC fate decisions as well as to develop robust directed lineage specification protocols.


Subject(s)
Embryonic Stem Cells/physiology , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Line , Computer Simulation , Gene Expression Profiling , Gene Knockdown Techniques , Mice , Reproducibility of Results , Systems Biology
11.
bioRxiv ; 2023 Sep 02.
Article in English | MEDLINE | ID: mdl-37425707

ABSTRACT

Cellular heterogeneity within the sinoatrial node (SAN) is functionally important but has been difficult to model in vitro , presenting a major obstacle to studies of heart rate regulation and arrhythmias. Here we describe a scalable method to derive sinoatrial node pacemaker cardiomyocytes (PCs) from human induced pluripotent stem cells that recapitulates differentiation into distinct PC subtypes, including SAN Head, SAN Tail, transitional zone cells, and sinus venosus myocardium. Single cell (sc) RNA-sequencing, sc-ATAC-sequencing, and trajectory analyses were used to define epigenetic and transcriptomic signatures of each cell type, and to identify novel transcriptional pathways important for PC subtype differentiation. Integration of our multi-omics datasets with genome wide association studies uncovered cell type-specific regulatory elements that associated with heart rate regulation and susceptibility to atrial fibrillation. Taken together, these datasets validate a novel, robust, and realistic in vitro platform that will enable deeper mechanistic exploration of human cardiac automaticity and arrhythmia.

12.
Stem Cells ; 29(11): 1705-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21915945

ABSTRACT

Embryonic stem cells (ESCs) derived from preimplantation blastocysts have unique self-renewal and multilineage differentiation properties that are controlled by key components of a core regulatory network including Oct4, Sox2, and Nanog. Understanding molecular underpinnings of these properties requires identification and characterization of additional factors that act in conjunction with these key factors in ESCs. We have previously identified Zfp281, a Krüppel-like zinc finger transcription factor, as an interaction partner of Nanog. We now present detailed functional analyses of Zfp281 using a genetically ablated null allele in mouse ESCs. Our data show that while Zfp281 is dispensable for establishment and maintenance of ESCs, it is required for their proper differentiation in vitro. We performed microarray profiling in combination with previously published datasets of Zfp281 global target gene occupancy and found that Zfp281 mainly functions as a repressor to restrict expression of many stem cell pluripotency genes. In particular, we demonstrated that deletion of Zfp281 resulted in upregulation of Nanog at both the transcript and protein levels with concomitant compromised differentiation of ESCs during embryoid body culture. Chromatin immunoprecipitation experiments demonstrated that Zfp281 is required for Nanog binding to its own promoter, suggesting that Nanog-associated repressive complex(es) involving Zfp281 may fine-tune Nanog expression for pluripotency of ESCs.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nanog Homeobox Protein , Protein Binding , Transcription Factors/genetics
13.
Stem Cells ; 29(6): 964-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21563278

ABSTRACT

Reprogramming patient-specific somatic cells into induced pluripotent stem (iPS) cells has great potential to develop feasible regenerative therapies. However, several issues need to be resolved such as ease, efficiency, and safety of generation of iPS cells. Many different cell types have been reprogrammed, most conveniently even peripheral blood mononuclear cells. However, they typically require the enforced expression of several transcription factors, posing mutagenesis risks as exogenous genetic material. To reduce this risk, iPS cells were previously generated with Oct4 alone from rather inaccessible neural stem cells that endogenously express the remaining reprogramming factors and very recently from fibroblasts with Oct4 alone in combination with additional small molecules. Here, we exploit that dermal papilla (DP) cells from hair follicles in the skin express all but one reprogramming factors to show that these accessible cells can be reprogrammed into iPS cells with the single transcription factor Oct4 and without further manipulation. Reprogramming was already achieved after 3 weeks and with efficiencies similar to other cell types reprogrammed with four factors. Dermal papilla-derived iPS cells are comparable to embryonic stem cells with respect to morphology, gene expression, and pluripotency. We conclude that DP cells may represent a preferred cell type for reprogramming accessible cells with less manipulation and for ultimately establishing safe conditions in the future by replacing Oct4 with small molecules.


Subject(s)
Hair Follicle/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Cell Differentiation , Cloning, Molecular , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Epigenesis, Genetic , Female , Fertilization in Vitro , Gene Expression Profiling , Genome , Genomic Imprinting , Hair Follicle/metabolism , Male , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , RNA Interference , Recombinant Proteins/genetics , Transgenes , Transplantation Chimera
14.
Stem Cells ; 28(2): 221-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20014278

ABSTRACT

Direct reprogramming of somatic cells into induced pluripotent stem (iPS) cells by only four transcription factors (Oct4, Sox2, Klf4, and c-Myc) has great potential for tissue-specific regenerative therapies, eliminating the ethical issues surrounding the use of embryonic stem cells and the rejection problems of using non-autologous cells. The reprogramming efficiency generally is very low, however, and the problems surrounding the introduction of viral genetic material are only partially investigated. Recent efforts to reduce the number of virally expressed transcription factors succeeded at reprogramming neural stem cells into iPS cells by overexpressing Oct4 alone. However, the relative inaccessibility and difficulty of obtaining neural cells in humans remains to be resolved. Here we report that dermal papilla (DP) cells, which are specialized skin fibroblasts thought to instruct hair follicle stem cells, endogenously express high levels of Sox2 and c-Myc, and that these cells can be reprogrammed into iPS cells with only Oct4 and Klf4. Moreover, we show that DP cells are reprogrammed more efficiently than skin and embryonic fibroblasts. iPS cells derived from DP cells expressed pluripotency genes and differentiated into cells from all germ layers in vitro and widely contributed to chimeric mice in vivo, including the germline. Our work establishes DP cells as an easily accessible source to generate iPS cells with efficiency and with less genetic material. This opens up the possibility of streamlined generation of skin-derived, patient-specific pluripotent stem cells and of ultimately replacing the remaining two factors with small molecules for safe generation of transplantable cells.


Subject(s)
Cellular Reprogramming/physiology , Dermis/cytology , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/metabolism , Animals , Cells, Cultured , Cellular Reprogramming/genetics , Dermis/metabolism , Female , Fibroblasts/cytology , Gonadotropins, Equine , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors
16.
Stem Cells ; 27(12): 2979-91, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19785031

ABSTRACT

Little is known about the molecular mechanism(s) governing differentiation decisions in embryonic stem cells (ESCs). To identify factors critical for ESC lineage formation, we carried out a functional genetic screen for factors affecting Nanog promoter activity during mESC differentiation. We report that members of the PBAF chromatin remodeling complex, including Smarca4/Brg1, Smarcb1/Baf47, Smarcc1/Baf155, and Smarce1/Baf57, are required for the repression of Nanog and other self-renewal gene expression upon mouse ESC (mESC) differentiation. Knockdown of Smarcc1 or Smarce1 suppressed loss of Nanog expression in multiple forms of differentiation. This effect occurred in the absence of self-renewal factors normally required for Nanog expression (e.g., Oct4), possibly indicating that changes in chromatin structure, rather than loss of self-renewal gene transcription per se, trigger differentiation. Consistent with this notion, mechanistic studies demonstrated that expression of Smarcc1 is necessary for heterochromatin formation and chromatin compaction during differentiation. Collectively, our data reveal that Smarcc1 plays important roles in facilitating mESCs differentiation by coupling gene repression with global and local changes in chromatin structure.


Subject(s)
Cell Differentiation , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Cell Line , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Protein Binding , Transcription Factors/genetics
17.
Stem Cells ; 26(8): 2019-31, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467660

ABSTRACT

The Wnt signaling pathway is necessary both for maintaining undifferentiated stem cells and for directing their differentiation. In mouse embryonic stem cells (ESCs), Wnt signaling preferentially maintains "stemness" under certain permissive conditions. T-cell factor 3 (Tcf3) is a component of the Wnt signaling and a dominant downstream effector in ESCs. Despite the wealth of knowledge regarding the importance of Wnt signaling underlying stem cells functions, the precise mechanistic explanation by which the effects are mediated is unknown. In this study, we identified new regulatory targets of Tcf3 using a whole-genome approach and found that Tcf3 transcriptionally represses many genes important for maintaining pluripotency and self-renewal, as well as those involved in lineage commitment and stem cell differentiation. This effect is in part mediated by the corepressors transducin-like enhancer of split 2 and C-terminal Binding Protein (CtBP). Notably, Tcf3 binds to and represses the Oct4 promoter, and this repressive effect requires both the Groucho and CtBP interacting domains of Tcf3. Interestingly, we find that in mouse preimplantation development embryos, Tcf3 expression is coregulated with Oct4 and Nanog and becomes localized to the inner cell mass of the blastocyst. These data demonstrate an important role for Tcf3 in modulating the appropriate level of gene transcription in ESCs and during embryonic development. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Embryonic Stem Cells/cytology , TCF Transcription Factors/metabolism , TCF Transcription Factors/physiology , Alcohol Oxidoreductases/metabolism , Animals , Blastocyst/cytology , Cell Differentiation , Cell Lineage , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Genome , Humans , Mice , Octamer Transcription Factor-3/metabolism , Transcription Factor 7-Like 1 Protein , Transcription, Genetic , Wnt Proteins/metabolism
18.
Stem Cells ; 26(1): 17-29, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17916804

ABSTRACT

Hundreds of microRNAs (miRNAs) are expressed in mammalian cells, where they aid in modulating gene expression by mediating mRNA transcript cleavage and/or regulation of translation rate. Functional studies to date have demonstrated that several of these miRNAs are important during development. However, the role of miRNAs in the regulation of stem cell growth and differentiation is not well understood. We show herein that microRNA (miR)-134 levels are maximally elevated at day 4 after retinoic acid-induced differentiation or day 2 after N2B27-induced differentiation of mouse embryonic stem cells (mESCs), but this change is not observed during embryoid body differentiation. The elevation of miR-134 levels alone in mESCs enhances differentiation toward ectodermal lineages, an effect that is blocked by a miR-134 antagonist. The promotion of mESC differentiation by miR-134 is due, in part, to its direct translational attenuation of Nanog and LRH1, both of which are known positive regulators of Oct4/POU5F1 and mESC growth. Together, the data demonstrate that miR-134 alone can enhance the differentiation of mESCs to ectodermal lineages and establish a functional role for miR-134 in modulating mESC differentiation through its potential to target and regulate multiple mRNAs.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Profiling , Genetic Vectors , In Situ Hybridization , Mice , Nanog Homeobox Protein , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
19.
Cell Rep ; 28(6): 1400-1409.e4, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390555

ABSTRACT

A multitude of signals are coordinated to maintain self-renewal in embryonic stem cells (ESCs). To unravel the essential internal and external signals required for sustaining the ESC state, we expand upon a set of ESC pluripotency-associated phosphoregulators (PRs) identified previously by short hairpin RNA (shRNA) screening. In addition to the previously described Aurka, we identify 4 additional PRs (Bub1b, Chek1, Ppm1g, and Ppp2r1b) whose depletion compromises self-renewal and leads to consequent differentiation. Global gene expression profiling and computational analyses reveal that knockdown of the 5 PRs leads to DNA damage/genome instability, activating p53 and culminating in ESC differentiation. Similarly, depletion of genome integrity-associated genes involved in DNA replication and checkpoint, mRNA processing, and Charcot-Marie-Tooth disease lead to compromise of ESC self-renewal via an increase in p53 activity. Our studies demonstrate an essential link between genomic integrity and developmental cell fate regulation in ESCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/physiology , Genomic Instability , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Line , DNA Damage , Gene Expression Profiling , Genetic Complementation Test , Mice , Phosphoproteins/genetics , Phosphoproteins/physiology , RNA, Small Interfering , Signal Transduction , Tumor Suppressor Protein p53/physiology
20.
Mol Cell Biol ; 25(14): 6031-46, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988017

ABSTRACT

Embryonic stem cells (ESCs) are pluripotent cells that can either self-renew or differentiate into many cell types. Oct4 and Sox2 are transcription factors essential to the pluripotent and self-renewing phenotypes of ESCs. Both factors are upstream in the hierarchy of the transcription regulatory network and are partners in regulating several ESC-specific genes. In ESCs, Sox2 is transcriptionally regulated by an enhancer containing a composite sox-oct element that Oct4 and Sox2 bind in a combinatorial interaction. It has previously been shown that Pou5f1, the Oct4 gene, contains a distal enhancer imparting specific expression in both ESCs and preimplantation embryos. Here, we identify a composite sox-oct element within this enhancer and show that it is involved in Pou5f1 transcriptional activity in ESCs. In vitro experiments with ESC nuclear extracts demonstrate that Oct4 and Sox2 interact specifically with this regulatory element. More importantly, by chromatin immunoprecipitation assay, we establish that both Oct4 and Sox2 bind directly to the composite sox-oct elements in both Pou5f1 and Sox2 in living mouse and human ESCs. Specific knockdown of either Oct4 or Sox2 by RNA interference leads to the reduction of both genes' enhancer activities and endogenous expression levels in addition to ESC differentiation. Our data uncover a positive and potentially self-reinforcing regulatory loop that maintains Pou5f1 and Sox2 expression via the Oct4/Sox2 complex in pluripotent cells.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HMGB Proteins/genetics , HMGB Proteins/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Embryo, Mammalian/cytology , Enhancer Elements, Genetic/genetics , Humans , Mice , Molecular Sequence Data , Octamer Transcription Factor-3 , Promoter Regions, Genetic/genetics , RNA Interference , SOXB1 Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL