Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 148(1-2): 259-72, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22225612

ABSTRACT

Identification of the factors critical to the tumor-initiating cell (TIC) state may open new avenues in cancer therapy. Here we show that the metabolic enzyme glycine decarboxylase (GLDC) is critical for TICs in non-small cell lung cancer (NSCLC). TICs from primary NSCLC tumors express high levels of the oncogenic stem cell factor LIN28B and GLDC, which are both required for TIC growth and tumorigenesis. Overexpression of GLDC and other glycine/serine enzymes, but not catalytically inactive GLDC, promotes cellular transformation and tumorigenesis. We found that GLDC induces dramatic changes in glycolysis and glycine/serine metabolism, leading to changes in pyrimidine metabolism to regulate cancer cell proliferation. In the clinic, aberrant activation of GLDC correlates with poorer survival in lung cancer patients, and aberrant GLDC expression is observed in multiple cancer types. This link between glycine metabolism and tumorigenesis may provide novel targets for advancing anticancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic , Glycine Dehydrogenase (Decarboxylating)/metabolism , Lung Neoplasms/metabolism , Amino Acid Sequence , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Fetal Proteins/metabolism , Glycine/metabolism , Humans , Molecular Sequence Data , Neoplasms/enzymology , Neoplasms/genetics , RNA-Binding Proteins , Sequence Alignment , Serine/metabolism , Thermus thermophilus/enzymology , Transplantation, Heterologous
2.
Cell ; 147(3): 525-38, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036562

ABSTRACT

The extent of lung regeneration following catastrophic damage and the potential role of adult stem cells in such a process remains obscure. Sublethal infection of mice with an H1N1 influenza virus related to that of the 1918 pandemic triggers massive airway damage followed by apparent regeneration. We show here that p63-expressing stem cells in the bronchiolar epithelium undergo rapid proliferation after infection and radiate to interbronchiolar regions of alveolar ablation. Once there, these cells assemble into discrete, Krt5+ pods and initiate expression of markers typical of alveoli. Gene expression profiles of these pods suggest that they are intermediates in the reconstitution of the alveolar-capillary network eradicated by viral infection. The dynamics of this p63-expressing stem cell in lung regeneration mirrors our parallel finding that defined pedigrees of human distal airway stem cells assemble alveoli-like structures in vitro and suggests new therapeutic avenues to acute and chronic airway disease.


Subject(s)
Bronchi/cytology , Influenza A Virus, H1N1 Subtype , Influenza, Human/pathology , Lung/physiology , Pulmonary Alveoli/cytology , Respiratory Distress Syndrome/pathology , Stem Cells/cytology , Animals , Disease Models, Animal , Gene Expression Profiling , Humans , Lung/cytology , Lung/virology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/virology , Rats , Transcription Factors/genetics , Wound Healing
3.
Physiol Rev ; 95(1): 245-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25540144

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.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Mammals/embryology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Embryonic Development , Humans
4.
Nat Methods ; 14(12): 1205-1212, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106405

ABSTRACT

Multiple adult tissues are maintained by stem cells of restricted developmental potential which can only form a subset of lineages within the tissue. For instance, the two adult lung epithelial compartments (airways and alveoli) are separately maintained by distinct lineage-restricted stem cells. A challenge has been to obtain multipotent stem cells and/or progenitors that can generate all epithelial cell types of a given tissue. Here we show that mouse Sox9+ multipotent embryonic lung progenitors can be isolated and expanded long term in 3D culture. Cultured Sox9+ progenitors transcriptionally resemble their in vivo counterparts and generate both airway and alveolar cell types in vitro. Sox9+ progenitors that were transplanted into injured adult mouse lungs differentiated into all major airway and alveolar lineages in vivo in a region-appropriate fashion. We propose that a single expandable embryonic lung progenitor population with broader developmental competence may eventually be used as an alternative for region-restricted adult tissue stem cells in regenerative medicine.


Subject(s)
Lung/cytology , Multipotent Stem Cells/cytology , SOX9 Transcription Factor/genetics , Animals , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knock-In Techniques , Lung/embryology , Lung/growth & development , Lung/metabolism , Mice, Transgenic , Multipotent Stem Cells/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , SOX9 Transcription Factor/metabolism , Tissue Engineering
5.
J Am Chem Soc ; 141(37): 14673-14686, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31436967

ABSTRACT

Tumor initiating cells (TIC) are resistant to conventional anticancer therapy and associated with metastasis and relapse in cancer. Although various TIC markers and their antibodies have been proposed, it is limited to the use of antibodies for in vivo imaging or treatment of TIC. In this study, we discovered heme oxygenase 2 (HMOX2) as a novel biomarker for TIC and developed a selective small molecule probe TiNIR (tumor initiating cell probe with near infrared). TiNIR detects and enriches the functionally active TIC in human lung tumors, and through the photoacoustic property, TiNIR also visualizes lung TIC in the patient-derived xenograft (PDX) model. Furthermore, we demonstrate that TiNIR inhibits tumor growth by blocking the function of HMOX2, resulting in significantly increased survival rates of the cancer model mice. The novel therapeutic target HMOX2 and its fluorescent ligand TiNIR will open a new path for the molecular level of lung TIC diagnosis and treatment.


Subject(s)
Fluorescent Dyes/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Spectroscopy, Near-Infrared/methods , Animals , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Mice , Neoplastic Stem Cells/enzymology , Survival Rate , Xenograft Model Antitumor Assays
6.
Nature ; 499(7456): 92-6, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23748442

ABSTRACT

Stem cells and progenitors in many lineages undergo self-renewing divisions, but the extracellular and intracellular proteins that regulate this process are largely unknown. Glucocorticoids stimulate red blood cell formation by promoting self-renewal of early burst-forming unit-erythroid (BFU-E) progenitors. Here we show that the RNA-binding protein ZFP36L2 is a transcriptional target of the glucocorticoid receptor (GR) in BFU-Es and is required for BFU-E self-renewal. ZFP36L2 is normally downregulated during erythroid differentiation from the BFU-E stage, but its expression is maintained by all tested GR agonists that stimulate BFU-E self-renewal, and the GR binds to several potential enhancer regions of ZFP36L2. Knockdown of ZFP36L2 in cultured BFU-E cells did not affect the rate of cell division but disrupted glucocorticoid-induced BFU-E self-renewal, and knockdown of ZFP36L2 in transplanted erythroid progenitors prevented expansion of erythroid lineage progenitors normally seen following induction of anaemia by phenylhydrazine treatment. ZFP36L2 preferentially binds to messenger RNAs that are induced or maintained at high expression levels during terminal erythroid differentiation and negatively regulates their expression levels. ZFP36L2 therefore functions as part of a molecular switch promoting BFU-E self-renewal and a subsequent increase in the total numbers of colony-forming unit-erythroid (CFU-E) progenitors and erythroid cells that are generated.


Subject(s)
Cell Division , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Tristetraprolin/metabolism , Animals , Cell Count , Cell Division/drug effects , Cell Lineage , Down-Regulation , Erythropoiesis/genetics , Gene Knockdown Techniques , Glucocorticoids/pharmacology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Stress, Physiological , Tristetraprolin/deficiency , Tristetraprolin/genetics
7.
Genes Dev ; 25(2): 119-24, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21196494

ABSTRACT

Using RNA-seq technology, we found that the majority of microRNAs (miRNAs) present in CFU-E erythroid progenitors are down-regulated during terminal erythroid differentiation. Of the developmentally down-regulated miRNAs, ectopic overexpression of miR-191 blocks erythroid enucleation but has minor effects on proliferation and differentiation. We identified two erythroid-enriched and developmentally up-regulated genes, Riok3 and Mxi1, as direct targets of miR-191. Knockdown of either Riok3 or Mxi1 blocks enucleation, and either physiological overexpression of miR-191 or knockdown of Riok3 or Mxi1 blocks chromatin condensation. Thus, down-regulation of miR-191 is essential for erythroid chromatin condensation and enucleation by allowing up-regulation of Riok3 and Mxi1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Erythroblasts/cytology , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Erythroblasts/metabolism , Gene Knockdown Techniques , Mice , Tumor Suppressor Proteins/genetics , Up-Regulation
8.
J Immunol ; 196(12): 4935-46, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183569

ABSTRACT

T cells undergo homeostatic expansion and acquire an activated phenotype in lymphopenic microenvironments. Restoration of normal lymphocyte numbers typically re-establishes normal homeostasis, and proinflammatory cytokine production returns to baseline. Mice deficient in guanine nucleotide exchange factor RasGRP1 exhibit dysregulated homeostatic expansion, which manifests as lymphoproliferative disease with autoantibody production. Our previous work revealed that autoreactive B cells lacking RasGRP1 break tolerance early during development, as well as during germinal center responses, suggesting that T cell-independent and T cell-dependent mechanisms are responsible. Examination of whether a particular T cell subset is involved in the breach of B cell tolerance revealed increased Th17 cells in Rasgrp1-deficient mice relative to control mice. Rasgrp1-deficient mice lacking IL-17R had fewer germinal centers, and germinal centers that formed contained fewer autoreactive B cells, suggesting that IL-17 signaling is required for a break in B cell tolerance in germinal centers. Interestingly, a fraction of Th17 cells from Rasgrp1-deficient mice were CXCR5(+) and upregulated levels of CD278 coordinate with their appearance in germinal centers, all attributes of T follicular helper cells (Tfh17). To determine whether CD278-CD275 interactions were required for the development of Tfh17 cells and for autoantibody, Rasgrp1-deficient mice were crossed with CD275-deficient mice. Surprisingly, mice deficient in RasGRP1 and CD275 formed Tfh17 cells and germinal centers and produced similar titers of autoantibodies as mice deficient in only RasGRP1. Therefore, these studies suggest that requirements for Tfh cell development change in lymphopenia-associated autoimmune settings.


Subject(s)
Autoimmunity , Germinal Center/immunology , Inducible T-Cell Co-Stimulator Ligand/immunology , Interleukin-17/immunology , Lymphopenia/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , B-Lymphocytes/immunology , Germinal Center/cytology , Guanine Nucleotide Exchange Factors/deficiency , Homeostasis , Immune Tolerance/genetics , Inducible T-Cell Co-Stimulator Ligand/deficiency , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-17/biosynthesis , Mice , Receptors, CXCR5/genetics , Receptors, Interleukin-17/deficiency , Signal Transduction , Th17 Cells/immunology
9.
Angew Chem Int Ed Engl ; 57(11): 2851-2854, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29377425

ABSTRACT

Tumor initiating cells (TICs) have been implicated in clinical relapse and metastasis of a variety of epithelial cancers, including lung cancer. While efforts toward the development of specific probes for TIC detection and targeting are ongoing, a universal TIC probe has yet to be developed. We report the first TIC-specific fluorescent chemical probe, TiY, with identification of the molecular target as vimentin, a marker for epithelial-to-mesenchymal transition (EMT). TiY selectively stains TICs over differentiated tumor cells or normal cells, and facilitates the visualization and enrichment of functionally active TICs from patient tumors. At high concentration, TiY also shows anti-TIC activity with low toxicity to non-TICs. With the unexplored target vimentin, TiY shows potential as a first universal probe for TIC detection in different cancers.


Subject(s)
Fluorescent Dyes/chemistry , Neoplastic Stem Cells/pathology , Small Molecule Libraries/chemistry , Vimentin/analysis , Animals , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Lung Neoplasms/pathology , Mice
10.
Stem Cells ; 34(2): 277-87, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26676652

ABSTRACT

In recent years, the highly conserved promyelocytic leukemia zinc finger (PLZF, also known as ZBTB16, ZNF145) has attracted attention as a multifunctional transcription factor involved in major biological processes during development. As a transcription factor, PLZF shows tight regulation in its cell-type-specific and stage-specific expression patterns. Emerging evidence shows that PLZF regulates the balance of self-renewal and differentiation in stem cells. However, the gene regulatory network of PLZF is only beginning to be understood. In this review, we discuss the diverse functions of PLZF, in particular its role in self-renewal versus differentiation of stem cells. We also discuss the current state of knowledge on the gene regulatory network of PLZF, in conjunction with its upstream factors, post-translational modifications and binding cofactors for multiprotein complexes. This review aims to provide the reader with an in-depth understanding of the molecular mechanisms underlying PLZF and the potential applications in tissue regeneration.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Gene Expression Regulation/physiology , Kruppel-Like Transcription Factors/metabolism , Protein Processing, Post-Translational/physiology , Stem Cells/metabolism , Animals , Humans , Promyelocytic Leukemia Zinc Finger Protein , Stem Cells/cytology
11.
Genes Dev ; 23(7): 862-76, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293287

ABSTRACT

The p53 transcription factor is a key tumor suppressor and a central regulator of the stress response. To ensure a robust and precise response to cellular signals, p53 gene expression must be tightly regulated from the transcriptional to the post-translational levels. Computational predictions suggest that several microRNAs are involved in the post-transcriptional regulation of p53. Here we demonstrate that miR-125b, a brain-enriched microRNA, is a bona fide negative regulator of p53 in both zebrafish and humans. miR-125b-mediated down-regulation of p53 is strictly dependent on the binding of miR-125b to a microRNA response element in the 3' untranslated region of p53 mRNA. Overexpression of miR-125b represses the endogenous level of p53 protein and suppresses apoptosis in human neuroblastoma cells and human lung fibroblast cells. In contrast, knockdown of miR-125b elevates the level of p53 protein and induces apoptosis in human lung fibroblasts and in the zebrafish brain. This phenotype can be rescued significantly by either an ablation of endogenous p53 function or ectopic expression of miR-125b in zebrafish. Interestingly, miR-125b is down-regulated when zebrafish embryos are treated with gamma-irradiation or camptothecin, corresponding to the rapid increase in p53 protein in response to DNA damage. Ectopic expression of miR-125b suppresses the increase of p53 and stress-induced apoptosis. Together, our study demonstrates that miR-125b is an important negative regulator of p53 and p53-induced apoptosis during development and during the stress response.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation , Genes, p53/physiology , MicroRNAs/metabolism , 3' Untranslated Regions/metabolism , Animals , Apoptosis/physiology , Base Sequence , Cell Line , Cell Line, Tumor , Cells, Cultured , Embryo, Nonmammalian/metabolism , Genetic Complementation Test , Humans , Molecular Sequence Data , Protein Binding , Stress, Physiological/physiology , Zebrafish
12.
Genes Dev ; 23(21): 2507-20, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19884257

ABSTRACT

The histone H3 Lys 9 (H3K9) methyltransferase Eset is an epigenetic regulator critical for the development of the inner cell mass (ICM). Although ICM-derived embryonic stem (ES) cells are normally unable to contribute to the trophectoderm (TE) in blastocysts, we find that depletion of Eset by shRNAs leads to differentiation with the formation of trophoblast-like cells and induction of trophoblast-associated gene expression. Using chromatin immmunoprecipitation (ChIP) and sequencing (ChIP-seq) analyses, we identified Eset target genes with Eset-dependent H3K9 trimethylation. We confirmed that genes that are preferentially expressed in the TE (Tcfap2a and Cdx2) are bound and repressed by Eset. Single-cell PCR analysis shows that the expression of Cdx2 and Tcfap2a is also induced in Eset-depleted morula cells. Importantly, Eset-depleted cells can incorporate into the TE of a blastocyst and, subsequently, placental tissues. Coimmunoprecipitation and ChIP assays further demonstrate that Eset interacts with Oct4, which in turn recruits Eset to silence these trophoblast-associated genes. Our results suggest that Eset restricts the extraembryonic trophoblast lineage potential of pluripotent cells and links an epigenetic regulator to key cell fate decision through a pluripotency factor.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Methyltransferases/metabolism , Octamer Transcription Factor-3/metabolism , Protein Methyltransferases/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , Chromatin Immunoprecipitation , Gene Expression Regulation, Developmental , Genome/physiology , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/metabolism , Mice , Morula/cytology , Transcription Factor AP-2/metabolism
13.
Nature ; 521(7552): 299-300, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25993958
14.
Nature ; 463(7284): 1096-100, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20139965

ABSTRACT

Induced pluripotent stem (iPS) cells can be obtained by the introduction of defined factors into somatic cells. The combination of Oct4 (also known as Pou5f1), Sox2 and Klf4 (which we term OSK) constitutes the minimal requirement for generating iPS cells from mouse embryonic fibroblasts. These cells are thought to resemble embryonic stem cells (ESCs) on the basis of global gene expression analyses; however, few studies have tested the ability and efficiency of iPS cells to contribute to chimaerism, colonization of germ tissues, and most importantly, germ-line transmission and live birth from iPS cells produced by tetraploid complementation. Using genomic analyses of ESC genes that have roles in pluripotency and fusion-mediated somatic cell reprogramming, here we show that the transcription factor Tbx3 significantly improves the quality of iPS cells. iPS cells generated with OSK and Tbx3 (OSKT) are superior in both germ-cell contribution to the gonads and germ-line transmission frequency. However, global gene expression profiling could not distinguish between OSK and OSKT iPS cells. Genome-wide chromatin immunoprecipitation sequencing analysis of Tbx3-binding sites in ESCs suggests that Tbx3 regulates pluripotency-associated and reprogramming factors, in addition to sharing many common downstream regulatory targets with Oct4, Sox2, Nanog and Smad1. This study underscores the intrinsic qualitative differences between iPS cells generated by different methods, and highlights the need to rigorously characterize iPS cells beyond in vitro studies.


Subject(s)
Chimera/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Gonads/cytology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Animals , Cell Fusion , Cellular Reprogramming , Chimera/embryology , Chromatin Immunoprecipitation , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Transgenic , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Regulatory Sequences, Nucleic Acid , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Smad1 Protein/metabolism , T-Box Domain Proteins/genetics , Transcription, Genetic/genetics , Transduction, Genetic
15.
Nature ; 468(7321): 316-20, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20953172

ABSTRACT

The derivation of human ES cells (hESCs) from human blastocysts represents one of the milestones in stem cell biology. The full potential of hESCs in research and clinical applications requires a detailed understanding of the genetic network that governs the unique properties of hESCs. Here, we report a genome-wide RNA interference screen to identify genes which regulate self-renewal and pluripotency properties in hESCs. Interestingly, functionally distinct complexes involved in transcriptional regulation and chromatin remodelling are among the factors identified in the screen. To understand the roles of these potential regulators of hESCs, we studied transcription factor PRDM14 to gain new insights into its functional roles in the regulation of pluripotency. We showed that PRDM14 regulates directly the expression of key pluripotency gene POU5F1 through its proximal enhancer. Genome-wide location profiling experiments revealed that PRDM14 colocalized extensively with other key transcription factors such as OCT4, NANOG and SOX2, indicating that PRDM14 is integrated into the core transcriptional regulatory network. More importantly, in a gain-of-function assay, we showed that PRDM14 is able to enhance the efficiency of reprogramming of human fibroblasts in conjunction with OCT4, SOX2 and KLF4. Altogether, our study uncovers a wealth of novel hESC regulators wherein PRDM14 exemplifies a key transcription factor required for the maintenance of hESC identity and the reacquisition of pluripotency in human somatic cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome, Human/genetics , RNA Interference , Repressor Proteins/metabolism , Animals , Base Sequence , Cell Line , Cellular Reprogramming/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , RNA-Binding Proteins , Repressor Proteins/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors
16.
Nucleic Acids Res ; 42(12): 7997-8007, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24860167

ABSTRACT

LIN28 function is fundamental to the activity and behavior of human embryonic stem cells (hESCs) and induced pluripotent stem cells. Its main roles in these cell types are the regulation of translational efficiency and let-7 miRNA maturation. However, LIN28-associated mRNA cargo shifting and resultant regulation of translational efficiency upon the initiation of differentiation remain unknown. An RNA-immunoprecipitation and microarray analysis protocol, eRIP, that has high specificity and sensitivity was developed to test endogenous LIN28-associated mRNA cargo shifting. A combined eRIP and polysome analysis of early stage differentiation of hESCs with two distinct differentiation cues revealed close similarities between the dynamics of LIN28 association and translational modulation of genes involved in the Wnt signaling, cell cycle, RNA metabolism and proteasomal pathways. Our data demonstrate that change in translational efficiency is a major contributor to early stages of differentiation of hESCs, in which LIN28 plays a central role. This implies that eRIP analysis of LIN28-associated RNA cargoes may be used for rapid functional quality control of pluripotent stem cells under manufacture for therapeutic applications.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Humans , Polyribosomes/metabolism
17.
J Immunol ; 191(7): 3605-13, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23997211

ABSTRACT

Lymphopenic hosts offer propitious microenvironments for expansion of autoreactive B and T cells. Despite this, many lymphopenic hosts do not develop autoimmune disease, suggesting that additional factors are required for breaching self-tolerance in the setting of lymphopenia. Mice deficient in guanine nucleotide exchange factor Rasgrp1 develop a lymphoproliferative disorder with features of human systemic lupus erythematosus. Early in life, Rasgrp1-deficient mice have normal B cell numbers but are T lymphopenic, leading to defective homeostatic expansion of CD4 T cells. To investigate whether B cell-intrinsic mechanisms also contribute to autoimmunity, Rasgrp1-deficient mice were bred to mice containing a knockin autoreactive BCR transgene (564Igi), thereby allowing the fate of autoreactive B cells to be assessed. During B cell development, the frequency of receptor-edited 564Igi B cells was reduced in Rasrp1-deficient mice compared with Rasgrp1-sufficient littermate control mice, suggesting that tolerance was impaired. In addition, the number of 564Igi transitional B cells was increased in Rasgrp1-deficient mice compared with control mice. Immature 564Igi B cells in bone marrow and spleen lacking RasGRP1 expressed lower levels of Bim mRNA and protein, suggesting that autoreactive B cells elude clonal deletion during development. Concomitant with increased serum autoantibodies, Rasgrp1-deficient mice developed spontaneous germinal centers at 8-10 wk of age. The frequency and number of 564Igi B cells within these germinal centers were significantly increased in Rasgrp1-deficient mice relative to control mice. Taken together, these studies suggest that autoreactive B cells lacking Rasgrp1 break central and peripheral tolerance through both T cell-independent and -dependent mechanisms.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Guanine Nucleotide Exchange Factors/genetics , Immune Tolerance/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Autoantibodies/blood , Autoantibodies/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Bcl-2-Like Protein 11 , Bone Marrow/immunology , Bone Marrow/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Guanine Nucleotide Exchange Factors/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Spleen/immunology , Spleen/metabolism , Toll-Like Receptors/metabolism
18.
J Immunol ; 191(6): 3192-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23935193

ABSTRACT

Engraftment of human CD34⁺ hematopoietic stem/progenitor cells into immunodeficient mice leads to robust reconstitution of human T and B cells but not monocytes and macrophages. To identify the cause underlying the poor monocyte and macrophage reconstitution, we analyzed human myeloid cell development in humanized mice and found that it was blocked at the promonocyte stage in the bone marrow. Expression of human M-CSF or GM-CSF by hydrodynamic injection of cytokine-encoding plasmid completely abolished the accumulation of promonocytes in the bone marrow. M-CSF promoted the development of mature monocytes and tissue-resident macrophages whereas GM-CSF did not. Moreover, correlating with an increased human macrophages at the sites of infection, M-CSF-treated humanized mice exhibited an enhanced protection against influenza virus and Mycobacterium infection. Our study identifies the precise stage at which human monocyte/macrophage development is blocked in humanized mice and reveals overlapping and distinct functions of M-CSF and GM-CSF in human monocyte and macrophage development. The improved reconstitution and functionality of monocytes/macrophages in the humanized mice following M-CSF expression provide a superior in vivo system to investigate the role of macrophages in physiological and pathological processes.


Subject(s)
Cell Differentiation/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Monocyte-Macrophage Precursor Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation/drug effects , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Monocyte-Macrophage Precursor Cells/immunology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Nat Genet ; 38(4): 431-40, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16518401

ABSTRACT

Oct4 and Nanog are transcription factors required to maintain the pluripotency and self-renewal of embryonic stem (ES) cells. Using the chromatin immunoprecipitation paired-end ditags method, we mapped the binding sites of these factors in the mouse ES cell genome. We identified 1,083 and 3,006 high-confidence binding sites for Oct4 and Nanog, respectively. Comparative location analyses indicated that Oct4 and Nanog overlap substantially in their targets, and they are bound to genes in different configurations. Using de novo motif discovery algorithms, we defined the cis-acting elements mediating their respective binding to genomic sites. By integrating RNA interference-mediated depletion of Oct4 and Nanog with microarray expression profiling, we demonstrated that these factors can activate or suppress transcription. We further showed that common core downstream targets are important to keep ES cells from differentiating. The emerging picture is one in which Oct4 and Nanog control a cascade of pathways that are intricately connected to govern pluripotency, self-renewal, genome surveillance and cell fate determination.


Subject(s)
DNA-Binding Proteins/physiology , Embryo, Mammalian/cytology , Homeodomain Proteins/physiology , Octamer Transcription Factor-3/physiology , Stem Cells/cytology , Transcription, Genetic/physiology , Animals , Embryo, Mammalian/metabolism , Gene Expression Regulation/physiology , Humans , Mice , Nanog Homeobox Protein , RNA Interference , Stem Cells/metabolism
20.
J Mol Cell Cardiol ; 67: 12-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24370890

ABSTRACT

The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.


Subject(s)
Cell Differentiation , Electrophysiological Phenomena , Myocytes, Cardiac/cytology , Culture Techniques , Epigenomics , Heart/embryology , Heart/growth & development , Humans , Pluripotent Stem Cells/cytology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL