Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 178(5): 1115-1131.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442404

ABSTRACT

Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (ßOHB), distinguishes self-renewing Lgr5+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes ßOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous ßOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, ßOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through ßOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of ßOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury.


Subject(s)
Diet, High-Fat , Ketone Bodies/metabolism , Stem Cells/metabolism , 3-Hydroxybutyric Acid/blood , 3-Hydroxybutyric Acid/pharmacology , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Cell Self Renewal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxymethylglutaryl-CoA Synthase/deficiency , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Intestines/cytology , Intestines/pathology , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Young Adult
2.
Cell ; 152(3): 570-83, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23352431

ABSTRACT

Long noncoding RNAs (lncRNAs) are often expressed in a development-specific manner, yet little is known about their roles in lineage commitment. Here, we identified Braveheart (Bvht), a heart-associated lncRNA in mouse. Using multiple embryonic stem cell (ESC) differentiation strategies, we show that Bvht is required for progression of nascent mesoderm toward a cardiac fate. We find that Bvht is necessary for activation of a core cardiovascular gene network and functions upstream of mesoderm posterior 1 (MesP1), a master regulator of a common multipotent cardiovascular progenitor. We also show that Bvht interacts with SUZ12, a component of polycomb-repressive complex 2 (PRC2), during cardiomyocyte differentiation, suggesting that Bvht mediates epigenetic regulation of cardiac commitment. Finally, we demonstrate a role for Bvht in maintaining cardiac fate in neonatal cardiomyocytes. Together, our work provides evidence for a long noncoding RNA with critical roles in the establishment of the cardiovascular lineage during mammalian development.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , RNA, Long Noncoding , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Regulatory Networks , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Myocytes, Cardiac/metabolism , Polycomb Repressive Complex 2/metabolism , Rats
3.
Cell ; 151(1): 206-20, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22981692

ABSTRACT

Heart development is exquisitely sensitive to the precise temporal regulation of thousands of genes that govern developmental decisions during differentiation. However, we currently lack a detailed understanding of how chromatin and gene expression patterns are coordinated during developmental transitions in the cardiac lineage. Here, we interrogated the transcriptome and several histone modifications across the genome during defined stages of cardiac differentiation. We find distinct chromatin patterns that are coordinated with stage-specific expression of functionally related genes, including many human disease-associated genes. Moreover, we discover a novel preactivation chromatin pattern at the promoters of genes associated with heart development and cardiac function. We further identify stage-specific distal enhancer elements and find enriched DNA binding motifs within these regions that predict sets of transcription factors that orchestrate cardiac differentiation. Together, these findings form a basis for understanding developmentally regulated chromatin transitions during lineage commitment and the molecular etiology of congenital heart disease.


Subject(s)
Epigenesis, Genetic , Gene Regulatory Networks , Myocardium/cytology , Animals , Cell Differentiation , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Heart/embryology , Humans , Mice , Transcription Factors/metabolism , Transcriptome
4.
Nature ; 585(7826): 603-608, 2020 09.
Article in English | MEDLINE | ID: mdl-32939090

ABSTRACT

Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.


Subject(s)
Ethers/metabolism , Ferroptosis , Peroxisomes/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Ethers/chemistry , Female , Gene Editing , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lipid Peroxidation , Male , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peroxisomes/genetics
5.
Mol Cell ; 64(1): 37-50, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27618485

ABSTRACT

Long non-coding RNAs (lncRNAs) are an emerging class of transcripts that can modulate gene expression; however, their mechanisms of action remain poorly understood. Here, we experimentally determine the secondary structure of Braveheart (Bvht) using chemical probing methods and show that this âˆ¼590 nt transcript has a modular fold. Using CRISPR/Cas9-mediated editing of mouse embryonic stem cells, we find that deletion of 11 nt in a 5' asymmetric G-rich internal loop (AGIL) of Bvht (bvhtdAGIL) dramatically impairs cardiomyocyte differentiation. We demonstrate a specific interaction between AGIL and cellular nucleic acid binding protein (CNBP/ZNF9), a zinc-finger protein known to bind single-stranded G-rich sequences. We further show that CNBP deletion partially rescues the bvhtdAGIL mutant phenotype by restoring differentiation capacity. Together, our work shows that Bvht functions with CNBP through a well-defined RNA motif to regulate cardiovascular lineage commitment, opening the door for exploring broader roles of RNA structure in development and disease.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Composition , Base Sequence , Binding Sites , CRISPR-Associated Protein 9 , Cell Differentiation , Cell Lineage/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/genetics , Endonucleases/metabolism , Gene Deletion , Gene Editing , Gene Expression Regulation , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Nucleic Acid Conformation , Phenotype , Protein Binding , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction
6.
Cell ; 135(4): 649-61, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18992931

ABSTRACT

Elucidating how chromatin influences gene expression patterns and ultimately cell fate is fundamental to understanding development and disease. The histone variant H2AZ has emerged as a key regulator of chromatin function and plays an essential but unknown role during mammalian development. Here, genome-wide analysis reveals that H2AZ occupies the promoters of developmentally important genes in a manner that is remarkably similar to that of the Polycomb group (PcG) protein Suz12. By using RNAi, we demonstrate a role for H2AZ in regulating target gene expression, find that H2AZ and PcG protein occupancy is interdependent at promoters, and further show that H2AZ is necessary for ES cell differentiation. Notably, H2AZ occupies a different subset of genes in lineage-committed cells, suggesting that its dynamic redistribution is necessary for cell fate transitions. Thus, H2AZ, together with PcG proteins, may establish specialized chromatin states in ES cells necessary for the proper execution of developmental gene expression programs.


Subject(s)
Embryonic Stem Cells/cytology , Histones/chemistry , Repressor Proteins/chemistry , Animals , Cell Differentiation , Cell Lineage , Chromatin/metabolism , Cluster Analysis , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Models, Biological , Polycomb-Group Proteins , Protein Binding , RNA Interference
7.
Development ; 146(19)2019 10 09.
Article in English | MEDLINE | ID: mdl-31427288

ABSTRACT

Deciphering the genetic and epigenetic regulation of cardiomyocyte proliferation in organisms that are capable of robust cardiac renewal, such as zebrafish, represents an attractive inroad towards regenerating the human heart. Using integrated high-throughput transcriptional and chromatin analyses, we have identified a strong association between H3K27me3 deposition and reduced sarcomere and cytoskeletal gene expression in proliferative cardiomyocytes following cardiac injury in zebrafish. To move beyond an association, we generated an inducible transgenic strain expressing a mutant version of histone 3, H3.3K27M, that inhibits H3K27me3 catalysis in cardiomyocytes during the regenerative window. Hearts comprising H3.3K27M-expressing cardiomyocytes fail to regenerate, with wound edge cells showing heightened expression of structural genes and prominent sarcomeres. Although cell cycle re-entry was unperturbed, cytokinesis and wound invasion were significantly compromised. Collectively, our study identifies H3K27me3-mediated silencing of structural genes as requisite for zebrafish heart regeneration and suggests that repression of similar structural components in the border zone of an infarcted human heart might improve its regenerative capacity.


Subject(s)
Gene Silencing , Heart/physiology , Histones/metabolism , Lysine/metabolism , Regeneration/physiology , Zebrafish/genetics , Zebrafish/physiology , Animals , Cell Proliferation , Cytokinesis , Cytoskeleton/metabolism , Gene Expression Regulation, Developmental , Methylation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism
8.
PLoS Genet ; 12(5): e1006034, 2016 05.
Article in English | MEDLINE | ID: mdl-27149122

ABSTRACT

Failure of the human heart to maintain sufficient output of blood for the demands of the body, heart failure, is a common condition with high mortality even with modern therapeutic alternatives. To identify molecular determinants of mortality in patients with new-onset heart failure, we performed a meta-analysis of genome-wide association studies and follow-up genotyping in independent populations. We identified and replicated an association for a genetic variant on chromosome 5q22 with 36% increased risk of death in subjects with heart failure (rs9885413, P = 2.7x10-9). We provide evidence from reporter gene assays, computational predictions and epigenomic marks that this polymorphism increases activity of an enhancer region active in multiple human tissues. The polymorphism was further reproducibly associated with a DNA methylation signature in whole blood (P = 4.5x10-40) that also associated with allergic sensitization and expression in blood of the cytokine TSLP (P = 1.1x10-4). Knockdown of the transcription factor predicted to bind the enhancer region (NHLH1) in a human cell line (HEK293) expressing NHLH1 resulted in lower TSLP expression. In addition, we observed evidence of recent positive selection acting on the risk allele in populations of African descent. Our findings provide novel genetic leads to factors that influence mortality in patients with heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Methylation/genetics , Heart Failure/genetics , Receptors, Cytokine/genetics , Black or African American/genetics , Alleles , Basic Helix-Loop-Helix Transcription Factors/blood , Chromosomes, Human, Pair 5/genetics , Female , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genotype , HEK293 Cells , Heart Failure/blood , Heart Failure/mortality , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Cytokine/blood
9.
Hum Mol Genet ; 25(10): 2093-2103, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962151

ABSTRACT

The ST-segment and adjacent T-wave (ST-T wave) amplitudes of the electrocardiogram are quantitative characteristics of cardiac repolarization. Repolarization abnormalities have been linked to ventricular arrhythmias and sudden cardiac death. We performed the first genome-wide association meta-analysis of ST-T-wave amplitudes in up to 37 977 individuals identifying 71 robust genotype-phenotype associations clustered within 28 independent loci. Fifty-four genes were prioritized as candidates underlying the phenotypes, including genes with established roles in the cardiac repolarization phase (SCN5A/SCN10A, KCND3, KCNB1, NOS1AP and HEY2) and others with as yet undefined cardiac function. These associations may provide insights in the spatiotemporal contribution of genetic variation influencing cardiac repolarization and provide novel leads for future functional follow-up.


Subject(s)
Arrhythmias, Cardiac/genetics , Brugada Syndrome/genetics , Electrocardiography , Genetic Predisposition to Disease , Genome-Wide Association Study , Adaptor Proteins, Signal Transducing/genetics , Arrhythmias, Cardiac/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Brugada Syndrome/physiopathology , Cardiac Conduction System Disease , Death, Sudden, Cardiac/pathology , Female , Heart Conduction System/physiopathology , Humans , Male , NAV1.5 Voltage-Gated Sodium Channel/genetics , Polymorphism, Single Nucleotide/genetics , Repressor Proteins/genetics , Shab Potassium Channels/genetics , Shal Potassium Channels/genetics
10.
EMBO J ; 32(13): 1805-16, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23756463

ABSTRACT

Cardiogenesis in mammals requires exquisite control of gene expression and faulty regulation of transcriptional programs underpins congenital heart disease (CHD), the most common defect among live births. Similarly, many adult cardiac diseases involve transcriptional changes and sometimes have a developmental basis. Long non-coding RNAs (lncRNAs) are a novel class of transcripts that regulate cellular processes by controlling gene expression; however, detailed insights into their biological and mechanistic functions are only beginning to emerge. Here, we discuss recent findings suggesting that lncRNAs are important factors in regulation of mammalian cardiogenesis and in the pathogenesis of CHD as well as adult cardiac disease. We also outline potential methodological and conceptual considerations for future studies of lncRNAs in the heart and other contexts.


Subject(s)
Gene Expression Regulation, Developmental , Heart Diseases/genetics , Transcription, Genetic , Adult , Heart Diseases/pathology , Humans , RNA, Long Noncoding
11.
Circ Res ; 117(2): 192-206, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26139858

ABSTRACT

Transcriptional and epigenetic regulation is critical for proper heart development, cardiac homeostasis, and pathogenesis. Long noncoding RNAs have emerged as key components of the transcriptional regulatory pathways that govern cardiac development as well as stress response, signaling, and remodeling in cardiac pathologies. Within the past few years, studies have identified many long noncoding RNAs in the context of cardiovascular biology and have begun to reveal the key functions of these transcripts. In this review, we discuss the growing roles of long noncoding RNAs in different aspects of cardiovascular development as well as pathological responses during injury or disease. In addition, we discuss diverse mechanisms by which long noncoding RNAs orchestrate cardiac transcriptional programs. Finally, we explore the exciting potential of this novel class of transcripts as biomarkers and novel therapeutic targets for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular Physiological Phenomena/genetics , Cardiovascular System/growth & development , Epigenesis, Genetic , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , Cardiovascular Diseases/physiopathology , Chromatin Assembly and Disassembly/physiology , Fetal Heart/metabolism , Forecasting , Gene Expression Regulation, Developmental , Heart/embryology , Heart Septal Defects, Ventricular/genetics , Histones/metabolism , Humans , Mice , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/classification
12.
Circ Res ; 116(5): 804-15, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25477501

ABSTRACT

RATIONALE: Neonatal mice have the capacity to regenerate their hearts in response to injury, but this potential is lost after the first week of life. The transcriptional changes that underpin mammalian cardiac regeneration have not been fully characterized at the molecular level. OBJECTIVE: The objectives of our study were to determine whether myocytes revert the transcriptional phenotype to a less differentiated state during regeneration and to systematically interrogate the transcriptional data to identify and validate potential regulators of this process. METHODS AND RESULTS: We derived a core transcriptional signature of injury-induced cardiac myocyte (CM) regeneration in mouse by comparing global transcriptional programs in a dynamic model of in vitro and in vivo CM differentiation, in vitro CM explant model, as well as a neonatal heart resection model. The regenerating mouse heart revealed a transcriptional reversion of CM differentiation processes, including reactivation of latent developmental programs similar to those observed during destabilization of a mature CM phenotype in the explant model. We identified potential upstream regulators of the core network, including interleukin 13, which induced CM cell cycle entry and STAT6/STAT3 signaling in vitro. We demonstrate that STAT3/periostin and STAT6 signaling are critical mediators of interleukin 13 signaling in CMs. These downstream signaling molecules are also modulated in the regenerating mouse heart. CONCLUSIONS: Our work reveals new insights into the transcriptional regulation of mammalian cardiac regeneration and provides the founding circuitry for identifying potential regulators for stimulating heart regeneration.


Subject(s)
Myocytes, Cardiac/metabolism , Regeneration/physiology , Transcription, Genetic , Animals , Animals, Newborn , Cell Adhesion Molecules/physiology , Cell Cycle , Cell Dedifferentiation/genetics , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , DNA Replication , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Heart Ventricles/cytology , Interleukin-13/pharmacology , Interleukin-13/physiology , Interleukin-13 Receptor alpha1 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-4 Receptor alpha Subunit/genetics , Mice , Muscle Development , Myocytes, Cardiac/drug effects , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/physiology , STAT6 Transcription Factor/physiology , Sequence Alignment , Transcription Factors/physiology , Transcriptome
13.
PLoS Genet ; 9(2): e1003288, 2013.
Article in English | MEDLINE | ID: mdl-23437007

ABSTRACT

SOX2 is a master regulator of both pluripotent embryonic stem cells (ESCs) and multipotent neural progenitor cells (NPCs); however, we currently lack a detailed understanding of how SOX2 controls these distinct stem cell populations. Here we show by genome-wide analysis that, while SOX2 bound to a distinct set of gene promoters in ESCs and NPCs, the majority of regions coincided with unique distal enhancer elements, important cis-acting regulators of tissue-specific gene expression programs. Notably, SOX2 bound the same consensus DNA motif in both cell types, suggesting that additional factors contribute to target specificity. We found that, similar to its association with OCT4 (Pou5f1) in ESCs, the related POU family member BRN2 (Pou3f2) co-occupied a large set of putative distal enhancers with SOX2 in NPCs. Forced expression of BRN2 in ESCs led to functional recruitment of SOX2 to a subset of NPC-specific targets and to precocious differentiation toward a neural-like state. Further analysis of the bound sequences revealed differences in the distances of SOX and POU peaks in the two cell types and identified motifs for additional transcription factors. Together, these data suggest that SOX2 controls a larger network of genes than previously anticipated through binding of distal enhancers and that transitions in POU partner factors may control tissue-specific transcriptional programs. Our findings have important implications for understanding lineage specification and somatic cell reprogramming, where SOX2, OCT4, and BRN2 have been shown to be key factors.


Subject(s)
Embryonic Stem Cells , Enhancer Elements, Genetic , Nerve Tissue Proteins , Octamer Transcription Factor-3 , POU Domain Factors , SOXB1 Transcription Factors , Animals , Cell Differentiation/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nucleotide Motifs , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
14.
PLoS Genet ; 9(8): e1003725, 2013.
Article in English | MEDLINE | ID: mdl-23990805

ABSTRACT

The histone H2A variant H2A.Z is essential for embryonic development and for proper control of developmental gene expression programs in embryonic stem cells (ESCs). Divergent regions of amino acid sequence of H2A.Z likely determine its functional specialization compared to core histone H2A. For example, H2A.Z contains three divergent residues in the essential C-terminal acidic patch that reside on the surface of the histone octamer as an uninterrupted acidic patch domain; however, we know little about how these residues contribute to chromatin structure and function. Here, we show that the divergent amino acids Gly92, Asp97, and Ser98 in the H2A.Z C-terminal acidic patch (H2A.Z(AP3)) are critical for lineage commitment during ESC differentiation. H2A.Z is enriched at most H3K4me3 promoters in ESCs including poised, bivalent promoters that harbor both activating and repressive marks, H3K4me3 and H3K27me3 respectively. We found that while H2A.Z(AP3) interacted with its deposition complex and displayed a highly similar distribution pattern compared to wild-type H2A.Z, its enrichment levels were reduced at target promoters. Further analysis revealed that H2A.Z(AP3) was less tightly associated with chromatin, suggesting that the mutant is more dynamic. Notably, bivalent genes in H2A.Z(AP3) ESCs displayed significant changes in expression compared to active genes. Moreover, bivalent genes in H2A.Z(AP3) ESCs gained H3.3, a variant associated with higher nucleosome turnover, compared to wild-type H2A.Z. We next performed single cell imaging to measure H2A.Z dynamics. We found that H2A.Z(AP3) displayed higher mobility in chromatin compared to wild-type H2A.Z by fluorescent recovery after photobleaching (FRAP). Moreover, ESCs treated with the transcriptional inhibitor flavopiridol resulted in a decrease in the H2A.Z(AP3) mobile fraction and an increase in its occupancy at target genes indicating that the mutant can be properly incorporated into chromatin. Collectively, our work suggests that the divergent residues in the H2A.Z acidic patch comprise a unique domain that couples control of chromatin dynamics to the regulation of developmental gene expression patterns during lineage commitment.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Histones/genetics , Animals , Asparagine/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Glycine/genetics , Mice , Nucleosomes/genetics , Promoter Regions, Genetic , Serine/genetics
15.
Nature ; 462(7271): 358-62, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19924215

ABSTRACT

Molecular regulation of embryonic stem cell (ESC) fate involves a coordinated interaction between epigenetic, transcriptional and translational mechanisms. It is unclear how these different molecular regulatory mechanisms interact to regulate changes in stem cell fate. Here we present a dynamic systems-level study of cell fate change in murine ESCs following a well-defined perturbation. Global changes in histone acetylation, chromatin-bound RNA polymerase II, messenger RNA (mRNA), and nuclear protein levels were measured over 5 days after downregulation of Nanog, a key pluripotency regulator. Our data demonstrate how a single genetic perturbation leads to progressive widespread changes in several molecular regulatory layers, and provide a dynamic view of information flow in the epigenome, transcriptome and proteome. We observe that a large proportion of changes in nuclear protein levels are not accompanied by concordant changes in the expression of corresponding mRNAs, indicating important roles for translational and post-translational regulation of ESC fate. Gene-ontology analysis across different molecular layers indicates that although chromatin reconfiguration is important for altering cell fate, it is preceded by transcription-factor-mediated regulatory events. The temporal order of gene expression alterations shows the order of the regulatory network reconfiguration and offers further insight into the gene regulatory network. Our studies extend the conventional systems biology approach to include many molecular species, regulatory layers and temporal series, and underscore the complexity of the multilayer regulatory mechanisms responsible for changes in protein expression that determine stem cell fate.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Animals , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Proteome , Time Factors
16.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-36798312

ABSTRACT

Expansion microscopy (ExM) enables nanoscale imaging using a standard confocal microscope through the physical, isotropic expansion of fixed immunolabeled specimens. ExM is widely employed to image proteins, nucleic acids, and lipid membranes in single cells at nanoscale resolution; however, current methods cannot be performed in multi-well cell culture plates which limits the number of samples that can be processed simultaneously. We developed High-throughput Expansion Microscopy (HiExM), a robust platform that enables expansion microscopy of cells cultured in a standard 96-well plate. Our method enables consistent ~4.2x expansion within individual wells, across multiple wells, and between plates processed in parallel. We also demonstrate that HiExM can be combined with high-throughput confocal imaging platforms greatly improve the ease and scalability of image acquisition. As an example, we analyzed the effects of doxorubicin, a known cardiotoxic agent, in human cardiomyocytes (CMs) based on Hoechst signal intensity. We show a dose dependent effect on nuclear chromatin that is not observed in unexpanded CMs, suggesting that HiExM improves the detection of cellular phenotypes in response to drug treatment. Our method broadens the application of ExM as a tool for scalable super-resolution imaging in biological research applications.

17.
Am J Sports Med ; 52(2): 503-515, 2024 02.
Article in English | MEDLINE | ID: mdl-38186352

ABSTRACT

BACKGROUND: The functional heterogeneity of culture-expanded mesenchymal stem cells (MSCs) has hindered the clinical application of MSCs. Previous studies have shown that MSC subpopulations with superior chondrogenic capacity can be isolated using a spiral microfluidic device based on the principle of inertial cell focusing. HYPOTHESIS: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function will overcome the challenge of the functional heterogeneity of expanded MSCs and will significantly improve MSC-based cartilage repair. STUDY DESIGN: Controlled laboratory study. METHODS: A next-generation, fully automated multidimensional double spiral microfluidic device was designed to provide more refined and efficient isolation of MSC subpopulations based on size. Analysis of in vitro chondrogenic potential and RNA sequencing was performed on size-sorted MSC subpopulations. In vivo cartilage repair efficacy was demonstrated in an osteochondral injury model in 12-week-old rats. Defects were implanted with MSC subpopulations (n = 6 per group) and compared with those implanted with unsegregated MSCs (n = 6). Osteochondral repair was assessed at 6 and 12 weeks after surgery by histological, micro-computed tomography, and mechanical analysis. RESULTS: A chondrogenic MSC subpopulation was efficiently isolated using the multidimensional double spiral device. RNA sequencing revealed distinct transcriptomic profiles and identified differential gene expression between subpopulations. The delivery of a chondrogenic MSC subpopulation resulted in improved cartilage repair, as indicated by histological scoring, the compression modulus, and micro-computed tomography of the subchondral bone. CONCLUSION: We have established a rapid, label-free, and reliable microfluidic protocol for more efficient size-based enrichment of a chondrogenic MSC subpopulation. Our proof-of-concept in vivo study demonstrates the enhanced cartilage repair efficacy of these enriched chondrogenic MSCs. CLINICAL RELEVANCE: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function can overcome the challenge of the functional heterogeneity of expanded MSCs, resulting in significant improvement in MSC-based cartilage repair. The availability of such rapid, label-free enriched chondrogenic MSCs can enable better cell therapy products for cartilage repair with improved treatment outcomes.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Rats , Cartilage, Articular/surgery , Microfluidics , X-Ray Microtomography , Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis
18.
Proc Natl Acad Sci U S A ; 107(50): 21931-6, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106759

ABSTRACT

Developmental programs are controlled by transcription factors and chromatin regulators, which maintain specific gene expression programs through epigenetic modification of the genome. These regulatory events at enhancers contribute to the specific gene expression programs that determine cell state and the potential for differentiation into new cell types. Although enhancer elements are known to be associated with certain histone modifications and transcription factors, the relationship of these modifications to gene expression and developmental state has not been clearly defined. Here we interrogate the epigenetic landscape of enhancer elements in embryonic stem cells and several adult tissues in the mouse. We find that histone H3K27ac distinguishes active enhancers from inactive/poised enhancer elements containing H3K4me1 alone. This indicates that the amount of actively used enhancers is lower than previously anticipated. Furthermore, poised enhancer networks provide clues to unrealized developmental programs. Finally, we show that enhancers are reset during nuclear reprogramming.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Histones/genetics , Mice , Mice, Inbred C57BL
19.
Stem Cells Transl Med ; 12(5): 266-280, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36988042

ABSTRACT

Detection of cellular senescence is important quality analytics of cell therapy products, including mesenchymal stromal cells (MSCs). However, its detection is critically limited by the lack of specific markers and the destructive assays used to read out these markers. Here, we establish a rapid, live-cell assay for detecting senescent cells in heterogeneous mesenchymal stromal cell (MSC) cultures. We report that the T2 relaxation time measured by microscale Magnetic Resonance Relaxometry, which is related to intracellular iron accumulation, correlates strongly with senescence markers in MSC cultures under diverse conditions, including different passages and donors, size-sorted MSCs by inertial spiral microfluidic device, and drug-induced senescence. In addition, the live-cell and non-destructive method presented here has general applicability to other cells and tissues and can critically advance our understanding of cellular senescence.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells , Cell Proliferation , Cell- and Tissue-Based Therapy , Magnetic Resonance Spectroscopy , Cells, Cultured
20.
Stem Cell Res Ther ; 14(1): 259, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37726837

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) have broad potential as a cell therapy including for the treatment of drug-resistant inflammatory conditions with abnormal T cell proliferation such as graft-versus-host disease (GVHD). Clinical success, however, has been complicated by the heterogeneity of culture-expanded MSCs as well as donor variability. Here, we devise culture conditions that promote expansion of MSCs with enhanced immunomodulatory functions both in vitro and in animal models of GVHD. METHODS: Human bone marrow-derived MSCs were expanded at high-confluency (MSCHC) and low-confluency state (MSCLC). Their immunomodulatory properties were evaluated with in vitro co-culture assays based on suppression of activated T cell proliferation and secretion of pro-inflammatory cytokines from activated T cells. Metabolic state of these cells was determined, while RNA sequencing was performed to explore transcriptome of these MSCs. Ex vivo expanded MSCHC or MSCLC was injected into human peripheral blood mononuclear cells (PBMC)-induced GVHD mouse model to determine their in vivo therapeutic efficacy based on clinical grade scoring, human CD45+ blood count and histopathological examination. RESULTS: As compared to MSCLC, MSCHC significantly reduced both the proliferation of anti-CD3/CD28-activated T cells and secretion of pro-inflammatory cytokines upon MSCHC co-culture across several donors even in the absence of cytokine priming. Mechanistically, metabolic analysis of MSCHC prior to co-culture with activated T cells showed increased glycolytic metabolism and lactate secretion compared to MSCLC, consistent with their ability to inhibit T cell proliferation. Transcriptome analysis further revealed differential expression of immunomodulatory genes including TRIM29, BPIFB4, MMP3 and SPP1 in MSCHC as well as enriched pathways including cytokine-cytokine receptor interactions, cell adhesion and PI3K-AKT signalling. Lastly, we demonstrate in a human PBMC-induced GVHD mouse model that delivery of MSCHC showed greater suppression of inflammation and improved outcomes compared to MSCLC and saline controls. CONCLUSION: Our study provides evidence that ex vivo expansion of MSCs at high confluency alters the metabolic and transcriptomic states of these cells. Importantly, this approach maximizes the production of MSCs with enhanced immunomodulatory functions without priming, thus providing a non-invasive and generalizable strategy for improving the use of MSCs for the treatment of inflammatory diseases.


Subject(s)
Leukocytes, Mononuclear , Mesenchymal Stem Cells , Animals , Mice , Humans , Bone Marrow , Phosphatidylinositol 3-Kinases , Cytokines , Disease Models, Animal , DNA-Binding Proteins , Transcription Factors , Intercellular Signaling Peptides and Proteins
SELECTION OF CITATIONS
SEARCH DETAIL