Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38559095

ABSTRACT

Pulmonary fibrosis is a devastating disease with no effective treatments to cure, stop or reverse the unremitting, fatal fibrosis. A critical barrier to treating this disease is the lack of understanding of the pathways leading to fibrosis as well as those regulating the resolution of fibrosis. Fibrosis is the pathologic side of normal tissue repair that results when the normal wound healing programs go awry. Successful resolution of tissue injury requires several highly coordinated pathways, and this research focuses on the interplay between these overlapping pathways: immune effectors, inflammatory mediators and fibroproliferation in the resolution of fibrosis. Previously we have successfully prevented, mitigated, and even reversed established fibrosis using vaccinia vaccination immunotherapy in two models of murine lung fibrosis. The mechanism by which vaccinia reverses fibrosis is by vaccine induced lung specific Th1 skewed tissue resident memory (TRMs) in the lung. In this study, we isolated a population of vaccine induced TRMs - CD49a+ CD4+ T cells - that are both necessary and sufficient to reverse established pulmonary fibrosis. Using adoptive cellular therapy, we demonstrate that intratracheal administration of CD49a+ CD4+ TRMs into established fibrosis, reverses the fibrosis histologically, by promoting a decrease in collagen, and functionally, by improving lung function, without the need for vaccination. Furthermore, co-culture of in vitro derived CD49+ CD4+ human TRMs with human fibroblasts from individuals with idiopathic pulmonary fibrosis (IPF) results in the down regulation of IPF fibroblast collagen production. Lastly, we demonstrate in human IPF lung histologic samples that CD49a+ CD4+ TRMs, which can down regulate human IPF fibroblast function, fail to increase in the IPF lungs, thus potentially failing to promote resolution. Thus, we define a novel unappreciated role for tissue resident memory T cells in regulating established lung fibrosis to promote resolution of fibrosis and re-establish lung homeostasis. We demonstrate that immunotherapy, in the form of adoptive transfer of CD49a+ CD4+ TRMs into the lungs of mice with established fibrosis, not only stops progression of the fibrosis but more importantly reverses the fibrosis. These studies provide the insight and preclinical rationale for a novel paradigm shifting approach of using cellular immunotherapy to treat lung fibrosis.

2.
J Exp Med ; 221(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38597954

ABSTRACT

Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Macrophages , Inflammation , RNA, Viral , Lung
3.
Nat Commun ; 13(1): 7137, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36414616

ABSTRACT

The lung's gas exchange surface is comprised of alveolar AT1 and AT2 cells that are corrupted in several common and deadly diseases. They arise from a bipotent progenitor whose differentiation is thought to be dictated by differential mechanical forces. Here we show the critical determinant is FGF signaling. Fgfr2 is expressed in the developing progenitors in mouse then restricts to nascent AT2 cells and remains on throughout life. Its ligands are expressed in surrounding mesenchyme and can, in the absence of exogenous mechanical cues, induce progenitors to form alveolospheres with intermingled AT2 and AT1 cells. FGF signaling directly and cell autonomously specifies AT2 fate; progenitors lacking Fgfr2 in vitro and in vivo exclusively acquire AT1 fate. Fgfr2 loss in AT2 cells perinatally results in reprogramming to AT1 identity, whereas loss or inhibition later in life triggers AT2 apoptosis and compensatory regeneration. We propose that Fgfr2 signaling selects AT2 fate during development, induces a cell non-autonomous AT1 differentiation signal, then continuously maintains AT2 identity and survival throughout life.


Subject(s)
Alveolar Epithelial Cells , Mesoderm , Animals , Mice , Cell Differentiation , Signal Transduction , Apoptosis
4.
Science ; 376(6594): eabl4896, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549404

ABSTRACT

Molecular characterization of cell types using single-cell transcriptome sequencing is revolutionizing cell biology and enabling new insights into the physiology of human organs. We created a human reference atlas comprising nearly 500,000 cells from 24 different tissues and organs, many from the same donor. This atlas enabled molecular characterization of more than 400 cell types, their distribution across tissues, and tissue-specific variation in gene expression. Using multiple tissues from a single donor enabled identification of the clonal distribution of T cells between tissues, identification of the tissue-specific mutation rate in B cells, and analysis of the cell cycle state and proliferative potential of shared cell types across tissues. Cell type-specific RNA splicing was discovered and analyzed across tissues within an individual.


Subject(s)
Atlases as Topic , Cells , Organ Specificity , RNA Splicing , Single-Cell Analysis , Transcriptome , B-Lymphocytes/metabolism , Cells/metabolism , Humans , Organ Specificity/genetics , T-Lymphocytes/metabolism
5.
Nat Med ; 27(3): 546-559, 2021 03.
Article in English | MEDLINE | ID: mdl-33654293

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2+TMPRSS2+ cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.


Subject(s)
COVID-19/epidemiology , COVID-19/genetics , Host-Pathogen Interactions/genetics , SARS-CoV-2/physiology , Sequence Analysis, RNA/statistics & numerical data , Single-Cell Analysis/statistics & numerical data , Virus Internalization , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/pathology , COVID-19/virology , Cathepsin L/genetics , Cathepsin L/metabolism , Datasets as Topic/statistics & numerical data , Demography , Female , Gene Expression Profiling/statistics & numerical data , Humans , Lung/metabolism , Lung/virology , Male , Middle Aged , Organ Specificity/genetics , Respiratory System/metabolism , Respiratory System/virology , Sequence Analysis, RNA/methods , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Single-Cell Analysis/methods
6.
Nature ; 587(7835): 619-625, 2020 11.
Article in English | MEDLINE | ID: mdl-33208946

ABSTRACT

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Subject(s)
Cells/classification , Cells/metabolism , Immunity , Lung/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Aged , Animals , Atlases as Topic , Biomarkers , Cell Communication , Cells/immunology , Chemokines/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Humans , Lung/immunology , Male , Mice , Middle Aged , Receptors, Lymphocyte Homing/metabolism , Signal Transduction , Stromal Cells/metabolism
7.
Nature ; 586(7831): 785-789, 2020 10.
Article in English | MEDLINE | ID: mdl-33057196

ABSTRACT

In the mammalian lung, an apparently homogenous mesh of capillary vessels surrounds each alveolus, forming the vast respiratory surface across which oxygen transfers to the blood1. Here we use single-cell analysis to elucidate the cell types, development, renewal and evolution of the alveolar capillary endothelium. We show that alveolar capillaries are mosaics; similar to the epithelium that lines the alveolus, the alveolar endothelium is made up of two intermingled cell types, with complex 'Swiss-cheese'-like morphologies and distinct functions. The first cell type, which we term the 'aerocyte', is specialized for gas exchange and the trafficking of leukocytes, and is unique to the lung. The other cell type, termed gCap ('general' capillary), is specialized to regulate vasomotor tone, and functions as a stem/progenitor cell in capillary homeostasis and repair. The two cell types develop from bipotent progenitors, mature gradually and are affected differently in disease and during ageing. This cell-type specialization is conserved between mouse and human lungs but is not found in alligator or turtle lungs, suggesting it arose during the evolution of the mammalian lung. The discovery of cell type specialization in alveolar capillaries transforms our understanding of the structure, function, regulation and maintenance of the air-blood barrier and gas exchange in health, disease and evolution.


Subject(s)
Capillaries/cytology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/cytology , Pulmonary Gas Exchange , Aging , Alligators and Crocodiles/anatomy & histology , Animals , Biological Evolution , Capillaries/metabolism , Cell Division , Cell Self Renewal , Cellular Senescence , Humans , Male , Mice , Pulmonary Alveoli/metabolism , Stem Cells/classification , Stem Cells/cytology , Turtles/anatomy & histology
8.
Nature ; 541(7636): 176-181, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28002412

ABSTRACT

Respiratory dysfunction is a notorious cause of perinatal mortality in infants and sleep apnoea in adults, but the mechanisms of respiratory control are not clearly understood. Mechanical signals transduced by airway-innervating sensory neurons control respiration; however, the physiological significance and molecular mechanisms of these signals remain obscured. Here we show that global and sensory neuron-specific ablation of the mechanically activated ion channel Piezo2 causes respiratory distress and death in newborn mice. Optogenetic activation of Piezo2+ vagal sensory neurons causes apnoea in adult mice. Moreover, induced ablation of Piezo2 in sensory neurons of adult mice causes decreased neuronal responses to lung inflation, an impaired Hering-Breuer mechanoreflex, and increased tidal volume under normal conditions. These phenotypes are reproduced in mice lacking Piezo2 in the nodose ganglion. Our data suggest that Piezo2 is an airway stretch sensor and that Piezo2-mediated mechanotransduction within various airway-innervating sensory neurons is critical for establishing efficient respiration at birth and maintaining normal breathing in adults.


Subject(s)
Apnea/physiopathology , Ion Channels/metabolism , Lung/physiology , Lung/physiopathology , Mechanotransduction, Cellular/physiology , Reflex/physiology , Animals , Animals, Newborn , Apnea/genetics , Death , Female , Ion Channels/deficiency , Ion Channels/genetics , Male , Mechanotransduction, Cellular/genetics , Mice , Nodose Ganglion/metabolism , Reflex/genetics , Respiration , Sensory Receptor Cells/metabolism , Tidal Volume
9.
Nat Genet ; 48(5): 537-43, 2016 05.
Article in English | MEDLINE | ID: mdl-27019112

ABSTRACT

The hepatic vasculature is essential for liver development, homeostasis and regeneration, yet the developmental program of hepatic vessel formation and the embryonic origin of the liver vasculature remain unknown. Here we show in mouse that endocardial cells form a primitive vascular plexus surrounding the liver bud and subsequently contribute to a substantial portion of the liver vasculature. Using intersectional genetics, we demonstrate that the endocardium of the sinus venosus is a source for the hepatic plexus. Inhibition of endocardial angiogenesis results in reduced endocardial contribution to the liver vasculature and defects in liver organogenesis. We conclude that a substantial portion of liver vessels derives from the endocardium and shares a common developmental origin with coronary arteries.


Subject(s)
Cell Lineage , Endocardium/embryology , Liver/embryology , Animals , Coronary Vessels/embryology , Endocardium/cytology , Endocardium/metabolism , Heart Atria/embryology , Heart Atria/metabolism , Heart Ventricles/embryology , Heart Ventricles/metabolism , Liver/blood supply , Liver Circulation , Mice , Morphogenesis , NFATC Transcription Factors/metabolism , Neovascularization, Physiologic
10.
Nat Med ; 21(8): 866-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168292

ABSTRACT

Unraveling the fate specification of resident stem cells during lung regeneration is of clinical importance. It has been reported that c-kit(+) progenitor cells resident in the human lung regenerate epithelial lineages upon transplantation into injured mouse lung. Here we test the lineage potential of c-kit(+) cells by inducible genetic lineage tracing. We find that c-kit(+) cells do not contribute to lung epithelium during homeostasis and repair, and instead maintain a vascular endothelial cell fate. These findings call attention to the clinical application of c-kit(+) stem cells as lung epithelial progenitors for the treatment of pulmonary disease.


Subject(s)
Endothelial Cells/physiology , Epithelial Cells/physiology , Lung/cytology , Proto-Oncogene Proteins c-kit/analysis , Animals , Cell Lineage , Female , Homeostasis , Male , Mice , Stem Cells/physiology
11.
Circulation ; 130(11 Suppl 1): S77-86, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25200059

ABSTRACT

BACKGROUND: Cell therapies offer the potential to improve cardiac function after myocardial infarction. Although injection of single-cell suspensions has proven safe, cell retention and survival rates are low. Tissue-engineered grafts allow cell delivery with minimal initial cell loss and mechanical support to the heart. However, graft performance cannot be easily compared, and optimal construct thickness, vascularization, and survival kinetics are unknown. METHODS AND RESULTS: Cardiac tissue slices (CTS) were generated by sectioning mouse hearts (n=40) expressing firefly luciferase and green fluorescent protein into slices of defined size and thickness using a vibrating blade microtome. Bioluminescence imaging of CTS transplanted onto hearts of immunodeficient mice demonstrated survival of ≤30% of transplanted cells. Cardiac slice perfusion was re-established within 3 days, likely through anastomosis of pre-existing vessels with the host vasculature and invasion of vessels from the host. Immunofluorescence showed a peak in cell death 3 days after transplantation and a gradual decline thereafter. MRI revealed preservation of contractile function and an improved ejection fraction 1 month after transplantation of CTS (28±2% CTS versus 22±2% control; P=0.05). Importantly, this effect was specific to CTS because transplantation of skeletal muscle tissue slices led to faster dilative remodeling and higher animal mortality. CONCLUSIONS: In summary, this is the first study to use CTS as a benchmark to validate and model tissue-engineered graft studies. CTS transplantation improved cell survival, established reperfusion, and enhanced cardiac function after myocardial infarction. These findings also confirm that dilative remodeling can be attenuated by topical transplantation of CTS but not skeletal muscle tissue grafts.


Subject(s)
Embryonic Stem Cells/transplantation , Heart Ventricles/transplantation , Myocardial Infarction/surgery , Tissue Engineering , Tissue Transplantation/methods , Animals , Animals, Newborn , Female , Genes, Reporter , Graft Survival , Green Fluorescent Proteins/genetics , Humans , Luciferases, Firefly/genetics , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Models, Animal , Myocardial Contraction , Organ Size , Quadriceps Muscle , Random Allocation
12.
Methods Mol Biol ; 1074: 15-29, 2013.
Article in English | MEDLINE | ID: mdl-23975802

ABSTRACT

Mouse postimplantation epiblast cultured in activin and basic fibroblast growth factor gives rise to continuously growing epiblast stem cells (EpiSCs) that share key properties with postimplantation epiblast, such as DNA methylation and an inactive X-chromosome. EpiSCs also show a distinct gene expression profile compared to embryonic stem cells (ESCs) derived from preimplantation blastocysts, and do not contribute efficiently to chimeras. EpiSCs can, however, revert to pluripotent ESC-like cells upon exposure to leukemia inhibitory factor-Stat3 signalling on feeder cells. Here we describe a protocol for the establishment of EpiSCs and their reversion to ESCs.


Subject(s)
Blastocyst/cytology , Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Germ Layers/cytology , Activins/pharmacology , Animals , DNA Methylation/genetics , Fibroblasts/cytology , Mice , Pluripotent Stem Cells/cytology , Signal Transduction
13.
J Cell Sci ; 125(Pt 24): 6094-104, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23077180

ABSTRACT

How cell fate becomes restricted during somatic cell differentiation is a long-lasting question in biology. Epigenetic mechanisms not present in pluripotent cells and acquired during embryonic development are expected to stabilize the differentiated state of somatic cells and thereby restrict their ability to convert to another fate. The histone variant macroH2A acts as a component of an epigenetic multilayer that heritably maintains the silent X chromosome and has been shown to restrict tumor development. Here we show that macroH2A marks the differentiated cell state during mouse embryogenesis. MacroH2A.1 was found to be present at low levels upon the establishment of pluripotency in the inner cell mass and epiblast, but it was highly enriched in the trophectoderm and differentiated somatic cells later in mouse development. Chromatin immunoprecipitation revealed that macroH2A.1 is incorporated in the chromatin of regulatory regions of pluripotency genes in somatic cells such as mouse embryonic fibroblasts and adult neural stem cells, but not in embryonic stem cells. Removal of macroH2A.1, macroH2A.2 or both increased the efficiency of induced pluripotency up to 25-fold. The obtained induced pluripotent stem cells reactivated pluripotency genes, silenced retroviral transgenes and contributed to chimeras. In addition, overexpression of macroH2A isoforms prevented efficient reprogramming of epiblast stem cells to naïve pluripotency. In summary, our study identifies for the first time a link between an epigenetic mark and cell fate restriction during somatic cell differentiation, which helps to maintain cell identity and antagonizes induction of a pluripotent stem cell state.


Subject(s)
Embryonic Stem Cells/physiology , Histones/metabolism , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cellular Reprogramming , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenomics , Female , Gene Expression Regulation, Developmental , Histones/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transfection
14.
Cell Stem Cell ; 11(1): 110-7, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770244

ABSTRACT

Blimp1 (Prdm1), the key determinant of primordial germ cells (PGCs), plays a combinatorial role with Prdm14 during PGC specification from postimplantation epiblast cells. They together initiate epigenetic reprogramming in early germ cells toward an underlying pluripotent state, which is equivalent to embryonic stem cells (ESCs). Whereas Prdm14 alone can promote reprogramming and is important for the propagation of the pluripotent state, it is not known whether Blimp1 is similarly involved. By using a genetic approach, we demonstrate that Blimp1 is dispensable for the derivation and maintenance of ESCs and postimplantation epiblast stem cells (epiSCs). Notably, Blimp1 is also dispensable for reprogramming epiSCs to ESCs. Thus, although Blimp1 is obligatory for PGC specification, it is not required for the reversion of epiSCs to ESCs and for their maintenance thereafter. This study suggests that reprogramming, including that of somatic cells to ESCs, may not entail an obligatory route through a Blimp1-positive PGC-like state.


Subject(s)
Germ Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cellular Reprogramming , Embryo Implantation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Germ Cells/cytology , Germ Layers/cytology , Germ Layers/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Positive Regulatory Domain I-Binding Factor 1 , Transcription Factors/deficiency
15.
Curr Opin Genet Dev ; 22(5): 466-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22795169

ABSTRACT

Development of mammalian primordial germ cells (PGCs) presents a unique example of a cell fate specification event that is intimately linked with epigenetic reprogramming. Cell fate commitment is governed by transcription factors which, together with epigenetic regulators, instruct lineage choice in response to signalling cues. Similarly, the reversal of epigenetic silencing is driven by the combinatorial action of transcriptional regulators, resulting in an increase in cellular plasticity. PGCs constitute a paradox, since their development as a unipotent specialised lineage is coupled with extensive reprogramming, which eventually leads to an increase in cellular potency. In this review we discuss the role of key factors in the specification of the germ cell lineage that are also important for the comprehensive erasure of epigenetic modifications, which provides the foundation for regeneration of totipotency. We further discuss current concepts of transcriptional and epigenetic control of cell fate decisions, with a particular focus on emerging principles of enhancer activity and their potential implications for the transcriptional control of PGC specification.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryonic Stem Cells/cytology , Germ Cells/metabolism , Animals , Cell Lineage , DNA Methylation , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Epigenomics , Gene Silencing , Humans , Mammals , Signal Transduction , Transcription Factors
16.
Cell Stem Cell ; 10(4): 425-39, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22482507

ABSTRACT

Epigenetic reprogramming in early germ cells is critical toward the establishment of totipotency, but investigations of the germline events are intractable. An objective cell culture-based system could provide mechanistic insight on how the key determinants of primordial germ cells (PGCs), including Prdm14, induce reprogramming in germ cells to an epigenetic ground state. Here we show a Prdm14-Klf2 synergistic effect that can accelerate and enhance reversion of mouse epiblast stem cells (epiSCs) to a naive pluripotent state, including X reactivation and DNA demethylation. Notably, Prdm14 alone has little effect on epiSC reversion, but it enhances the competence for reprogramming and potentially PGC specification. Reprogramming of epiSCs by the combinatorial effect of Prdm14-Klf2 involves key epigenetic changes, which might have an analogous role in PGCs. Our study provides a paradigm toward a systematic analysis of how other key genes contribute to complex and dynamic events of reprogramming in the germline.


Subject(s)
Cell Dedifferentiation , Epigenesis, Genetic , Germ Layers/metabolism , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , DNA Methylation , DNA-Binding Proteins , Germ Cells/cytology , Germ Layers/cytology , Humans , Mice , RNA-Binding Proteins , Stem Cells/cytology
17.
Nucleus ; 2(6): 533-9, 2011.
Article in English | MEDLINE | ID: mdl-22064467

ABSTRACT

How various epigenetic mechanisms restrict chromatin plasticity to determine the stability of repressed genes is poorly understood. Nuclear transfer to Xenopus oocytes induces the transcriptional reactivation of previously silenced genes. Recent work suggests that it can be used to analyze the epigenetic stability of repressed states. The notion that the epigenetic state of genes is an important determinant of the efficiency of nuclear reprogramming is supported by the differential reprogramming of given genes from different starting epigenetic configurations. After nuclear transfer, transcription from the inactive X chromosome of post-implantation-derived epiblast stem cells is reactivated. However, the same chromosome is resistant to reactivation when embryonic fibroblasts are used. Here, we discuss different kinds of evidence that link the histone variant macroH2A to the increased stability of repressed states. We focus on developmentally regulated X chromosome inactivation and repression of autosomal pluripotency genes, where macroH2A may help maintain the long-term stability of the differentiated state of somatic cells.


Subject(s)
Epigenesis, Genetic/physiology , Histones/metabolism , Nuclear Transfer Techniques , Oocytes/metabolism , Repressor Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Histones/genetics , Oocytes/cytology , Repressor Proteins/genetics , Transcription, Genetic/physiology , X Chromosome/genetics , X Chromosome/metabolism , Xenopus Proteins/genetics , Xenopus laevis
18.
EMBO J ; 30(12): 2373-87, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21552206

ABSTRACT

How various layers of epigenetic repression restrict somatic cell nuclear reprogramming is poorly understood. The transfer of mammalian somatic cell nuclei into Xenopus oocytes induces transcriptional reprogramming of previously repressed genes. Here, we address the mechanisms that restrict reprogramming following nuclear transfer by assessing the stability of the inactive X chromosome (Xi) in different stages of inactivation. We find that the Xi of mouse post-implantation-derived epiblast stem cells (EpiSCs) can be reversed by nuclear transfer, while the Xi of differentiated or extraembryonic cells is irreversible by nuclear transfer to oocytes. After nuclear transfer, Xist RNA is lost from chromatin of the Xi. Most epigenetic marks such as DNA methylation and Polycomb-deposited H3K27me3 do not explain the differences between reversible and irreversible Xi. Resistance to reprogramming is associated with incorporation of the histone variant macroH2A, which is retained on the Xi of differentiated cells, but absent from the Xi of EpiSCs. Our results uncover the decreased stability of the Xi in EpiSCs, and highlight the importance of combinatorial epigenetic repression involving macroH2A in restricting transcriptional reprogramming by oocytes.


Subject(s)
Cellular Reprogramming/genetics , Chromosomal Instability/genetics , Genetic Variation , Histones/genetics , Pluripotent Stem Cells/chemistry , RNA, Untranslated/chemistry , RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/chemistry , Male , Mice , Mice, Transgenic , Oocytes/chemistry , Oocytes/cytology , Oocytes/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding , Transcription, Genetic , Xenopus
19.
Differentiation ; 81(5): 281-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21330045

ABSTRACT

Depending on their origin, embryo-derived stem cells have distinct properties that largely correspond to their counterpart in vivo. Mouse epiblast stem cells derived from post-implantation embryos differ from embryonic stem cells derived from blastocysts in their transcriptional and epigenetic profile, their morphology and culture requirements. When maintained in appropriate conditions, the cells keep self-renewing and do not adopt a different state. Recent studies, however, show that it is possible to convert between stem cell states. Here we review recent advances to induce stem cell state changes and we consider the potential of germ cell-mediated reprogramming for the conversion. Since the properties of mouse epiblast stem cells are similar to human embryonic stem cells, we discuss the significance of stem cell conversion and germ cell-mediated reprogramming in humans.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Germ Cells/cytology , Germ Cells/metabolism , Germ Layers/metabolism , Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Epigenomics , Germ Layers/cytology , Humans , Mice , Stem Cells/metabolism
20.
Nature ; 461(7268): 1292-5, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19816418

ABSTRACT

The pluripotent state, which is first established in the primitive ectoderm cells of blastocysts, is lost progressively and irreversibly during subsequent development. For example, development of post-implantation epiblast cells from primitive ectoderm involves significant transcriptional and epigenetic changes, including DNA methylation and X chromosome inactivation, which create a robust epigenetic barrier and prevent their reversion to a primitive-ectoderm-like state. Epiblast cells are refractory to leukaemia inhibitory factor (LIF)-STAT3 signalling, but they respond to activin/basic fibroblast growth factor to form self-renewing epiblast stem cells (EpiSCs), which exhibit essential properties of epiblast cells and that differ from embryonic stem (ES) cells derived from primitive ectoderm. Here we show reprogramming of advanced epiblast cells from embryonic day 5.5-7.5 mouse embryos with uniform expression of N-cadherin and inactive X chromosome to ES-cell-like cells (rESCs) in response to LIF-STAT3 signalling. Cultured epiblast cells overcome the epigenetic barrier progressively as they proceed with the erasure of key properties of epiblast cells, resulting in DNA demethylation, X reactivation and expression of E-cadherin. The accompanying changes in the transcriptome result in a loss of phenotypic and epigenetic memory of epiblast cells. Using this approach, we report reversion of established EpiSCs to rESCs. Moreover, unlike epiblast and EpiSCs, rESCs contribute to somatic tissues and germ cells in chimaeras. Further studies may reveal how signalling-induced epigenetic reprogramming may promote reacquisition of pluripotency.


Subject(s)
Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Germ Layers/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cadherins/metabolism , Cells, Cultured , Cellular Reprogramming , DNA Methylation , Ectoderm/cytology , Embryo, Mammalian/cytology , Gene Expression Profiling , Germ Layers/metabolism , Leukemia Inhibitory Factor/metabolism , Mice , STAT3 Transcription Factor/metabolism , Y Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...