Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
2.
4.
Cell Stem Cell ; 21(1): 150, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28686865

ABSTRACT

This i3 is a data visualization based on the Cell Stem Cell tenth anniversary theme of lineage tracing. Using Scopus citations of Cell Stem Cell research papers, it illustrates both the evolution of the stem cell field and the way new research builds on work that came before. Users can navigate the graphic and the represented papers by stem cell type, organism, and author online at cell.com/i3/cell-stem-cell/lineage. To view this SnapShot, open or download the PDF.


Subject(s)
Biomedical Research , Stem Cells/metabolism , Animals , Humans , Periodicals as Topic , Stem Cells/cytology
5.
6.
Cell Stem Cell ; 18(2): 294, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26849306

ABSTRACT

Research into induced pluripotent stem cells (iPSCs) has expanded at a remarkable pace in the decade since Shinya Yamanaka and Kazutoshi Takahashi first reported their groundbreaking discovery in 2006. This Timeline highlights the key events in the development of this field, including basic insights into the production of iPSCs and how they have been applied to improve our understanding and treatment of human disease. To view this Timeline, open or download the PDF. You can also listen to the associated interview with Debbie Sweet, Editor of Cell Stem Cell, and Elena Porro, Editor of Cell. PAPERCLIP.


Subject(s)
Induced Pluripotent Stem Cells , Stem Cell Research/history , Cellular Reprogramming , History, 21st Century , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation
7.
Cell ; 164(3): 580, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824663

ABSTRACT

Research into induced pluripotent stem cells (iPSCs) has expanded at a remarkable pace in the decade since Shinya Yamanaka and Kazutoshi Takahashi first reported their groundbreaking discovery in 2006. This Timeline highlights the key events in the development of this field, including basic insights into the production of iPSCs and how they have been applied to improve our understanding and treatment of human disease.


Subject(s)
Induced Pluripotent Stem Cells , Stem Cell Research/history , Cell Culture Techniques , History, 21st Century , Humans , Stem Cell Transplantation
8.
Genome Res ; 24(2): 267-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24310002

ABSTRACT

Both diffusible factors acting in trans and chromatin components acting in cis are implicated in gene regulation, but the extent to which either process causally determines a cell's transcriptional identity is unclear. We recently used cell fusion to define a class of silent genes termed "cis-silenced" (or "occluded") genes, which remain silent even in the presence of trans-acting transcriptional activators. We further showed that occlusion of lineage-inappropriate genes plays a critical role in maintaining the transcriptional identities of somatic cells. Here, we present, for the first time, a comprehensive map of occluded genes in somatic cells. Specifically, we mapped occluded genes in mouse fibroblasts via fusion to a dozen different rat cell types followed by whole-transcriptome profiling. We found that occluded genes are highly prevalent and stable in somatic cells, representing a sizeable fraction of silent genes. Occluded genes are also highly enriched for important developmental regulators of alternative lineages, consistent with the role of occlusion in safeguarding cell identities. Alongside this map, we also present whole-genome maps of DNA methylation and eight other chromatin marks. These maps uncover a complex relationship between chromatin state and occlusion. Furthermore, we found that DNA methylation functions as the memory of occlusion in a subset of occluded genes, while histone deacetylation contributes to the implementation but not memory of occlusion. Our data suggest that the identities of individual cell types are defined largely by the occlusion status of their genomes. The comprehensive reference maps reported here provide the foundation for future studies aimed at understanding the role of occlusion in development and disease.


Subject(s)
Gene Expression Regulation , Gene Silencing , Regulatory Sequences, Nucleic Acid , Trans-Activators/genetics , Transcription, Genetic , Animals , Cell Fusion , Cell Line , Chromatin/genetics , DNA Methylation/genetics , Genome , Histones/genetics , Histones/metabolism , Mice , Rats
9.
Mol Cell ; 46(2): 159-70, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22445485

ABSTRACT

It is a long-held paradigm that cell fusion reprograms gene expression but the extent of reprogramming and whether it is affected by the cell types employed remain unknown. We recently showed that the silencing of somatic genes is attributable to either trans-acting cellular environment or cis-acting chromatin context. Here, we examine how trans- versus cis-silenced genes in a somatic cell type behave in fusions to another somatic cell type or to embryonic stem cells (ESCs). We demonstrate that while reprogramming of trans-silenced somatic genes occurs in both cases, reprogramming of cis-silenced somatic genes occurs only in somatic-ESC fusions. Importantly, ESCs reprogram the somatic genome in two distinct phases: trans-reprogramming occurs rapidly, independent of DNA replication, whereas cis-reprogramming occurs with slow kinetics requiring DNA replication. We also show that pluripotency genes Oct4 and Nanog are cis-silenced in somatic cells. We conclude that cis-reprogramming capacity is a fundamental feature distinguishing ESCs from somatic cells.


Subject(s)
Cell Fusion , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , DNA Replication , Gene Silencing , Kinetics , Mice
10.
J Immunol ; 185(1): 410-7, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20511547

ABSTRACT

Ikaros and Notch are transcriptional regulators essential for normal T cell development. Aberrant activation of Notch target genes is observed in Ikaros-deficient thymocytes as well as leukemia cell lines. However, it is not known whether Notch deregulation plays a preferential or obligatory role in the leukemia that arise in Ikaros null (Ik(-/-)) mice. To answer this question, the expression of the DNA-binding Notch target gene activator RBP-Jkappa was abrogated in Ik(-/-) double-positive thymocytes. This was accomplished through conditional inactivation using CD4-Cre transgenic mice containing floxed RBP-Jkappa alleles (RBPJ(fl/fl)). Ik(-/-) x RBPJ(fl/fl) x CD4-Cre(+) transgenic mice develop clonal T cell populations in the thymus that escape to the periphery, with similar kinetics and penetrance as their CD4-Cre(-) counterparts. The clonal populations do not display increased RBP-Jkappa expression compared with nontransformed thymocytes, suggesting there is no selection for clones that have not fully deleted RBP-Jkappa. However, RBPJ-deficient clonal populations do not expand as aggressively as their RBPJ-sufficient counterparts, suggesting a qualitative role for deregulated Notch target gene activation in the leukemogenic process. Finally, these studies show that RBP-Jkappa plays no role in Notch target gene repression in double-positive thymocytes but rather that it is Ikaros that is required for the repression of these genes at this critical stage of T cell development.


Subject(s)
Gene Targeting , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunophenotyping , Leukemia, Experimental/genetics , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Animals , Cell Line , Cell Line, Tumor , Gene Silencing/immunology , Ikaros Transcription Factor/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Notch/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
11.
J Immunol ; 181(9): 6265-74, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941217

ABSTRACT

Both Ikaros and Notch are essential for normal T cell development. Collaborative mutations causing a reduction in Ikaros activity and an increase in Notch activation promote T cell leukemogenesis. Although the molecular mechanisms of this cooperation have been studied, its consequences in thymocyte development remain unexplored. In this study, we show that Ikaros regulates expression of a subset of Notch target genes, including Hes1, Deltex1, pTa, Gata3, and Runx1, in both Ikaros null T cell leukemia lines and Ikaros null primary thymocytes. In Ikaros null leukemia cells, Notch deregulation occurs at both the level of Notch receptor cleavage and expression of Notch target genes, because re-expression of Ikaros in these cells down-regulates Notch target gene expression without affecting levels of intracellular cleaved Notch. In addition, abnormal expression of Notch target genes is observed in Ikaros null double-positive thymocytes, in the absence of detectable intracellular cleaved Notch. Finally, we show that this role of Ikaros is specific to double-positive and single-positive thymocytes because derepression of Notch target gene expression is not observed in Ikaros null double-negative thymocytes or lineage-depleted bone marrow. Thus, in this study, we provide evidence that Ikaros and Notch play opposing roles in regulation of a subset of Notch target genes and that this role is restricted to developing thymocytes where Ikaros is required to appropriately regulate the Notch program as they progress through T cell development.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation, Neoplastic/immunology , Ikaros Transcription Factor/physiology , Receptors, Notch/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Coculture Techniques , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Notch/biosynthesis , Receptors, Notch/genetics , Receptors, Notch/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/pathology , Transcription Factor HES-1 , Ubiquitin-Protein Ligases
12.
J Biol Chem ; 283(16): 10476-84, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18287091

ABSTRACT

Ikaros and Notch1, two regulators of gene transcription, are critically important at many stages of T cell development. Deregulation of Ikaros and Notch activities cooperate to promote T cell leukemogenesis, providing evidence that they function in converging pathways in developing T cells. In this report, a mechanism for Ikaros:Notch cooperativity is described, revealing a non-redundant role for Ikaros in regulating expression of the Notch target gene Hes1 in a leukemia T cell line. We provide evidence that Ikaros directly represses Hes1 in concert with the transcriptional repressor, RBP-Jkappa, allowing for cross-talk between Notch and Ikaros that impacts regulation of CD4 expression. Taken together, these data describe a potential mechanism for Ikaros' function during T cell development and define Ikaros as an obligate repressor of Hes1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4 Antigens/biosynthesis , Gene Expression Regulation , Homeodomain Proteins/metabolism , Ikaros Transcription Factor/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptor, Notch1/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Gene Silencing , Humans , Mice , Models, Biological , Molecular Sequence Data , Promoter Regions, Genetic , Transcription Factor HES-1
SELECTION OF CITATIONS
SEARCH DETAIL