Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Genome Res ; 29(6): 969-977, 2019 06.
Article in English | MEDLINE | ID: mdl-31160376

ABSTRACT

Chromatin features are characterized by genome-wide assays for nucleosome location, protein binding sites, three-dimensional interactions, and modifications to histones and DNA. For example, assay for transposase accessible chromatin sequencing (ATAC-seq) identifies nucleosome-depleted (open) chromatin, which harbors potentially active gene regulatory sequences; and bisulfite sequencing (BS-seq) quantifies DNA methylation. When two distinct chromatin features like these are assayed separately in populations of cells, it is impossible to determine, with certainty, where the features are coincident in the genome by simply overlaying data sets. Here, we describe methyl-ATAC-seq (mATAC-seq), which implements modifications to ATAC-seq, including subjecting the output to BS-seq. Merging these assays into a single protocol identifies the locations of open chromatin and reveals, unambiguously, the DNA methylation state of the underlying DNA. Such combinatorial methods eliminate the need to perform assays independently and infer where features are coincident.


Subject(s)
Chromatin/genetics , DNA Methylation , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , DNA Transposable Elements , DNA-Binding Proteins , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, DNA
2.
Methods ; 72: 41-50, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25204781

ABSTRACT

Dynamically regulated changes in chromatin states are vital for normal development and can produce disease when they go awry. Accordingly, much effort has been devoted to characterizing these states under normal and pathological conditions. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) is the most widely used method to characterize where in the genome transcription factors, modified histones, modified nucleotides and chromatin binding proteins are found; bisulfite sequencing (BS-seq) and its variants are commonly used to characterize the locations of DNA modifications. Though very powerful, these methods are not without limitations. Notably, they are best at characterizing one chromatin feature at a time, yet chromatin features arise and function in combination. Investigators commonly superimpose separate ChIP-seq or BS-seq datasets, and then infer where chromatin features are found together. While these inferences might be correct, they can be misleading when the chromatin source has distinct cell types, or when a given cell type exhibits any cell to cell variation in chromatin state. These ambiguities can be eliminated by robust methods that directly characterize the existence and genomic locations of combinations of chromatin features in very small inputs of cells or ideally, single cells. Here we review single molecule epigenomic methods under development to overcome these limitations, the technical challenges associated with single molecule methods and their potential application to single cells.


Subject(s)
Epigenomics/methods , Single-Cell Analysis/methods , Chromatin Immunoprecipitation/methods , DNA Methylation , Mass Spectrometry/methods , Microfluidics/methods , Software , Workflow
3.
Proc Natl Acad Sci U S A ; 110(19): 7772-7, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23610441

ABSTRACT

Proper placement of epigenetic marks on DNA and histones is fundamental to normal development, and perturbations contribute to a variety of disease states. Combinations of marks act together to control gene expression; therefore, detecting their colocalization is important, but because of technical challenges, such measurements are rarely reported. Instead, measurements of epigenetic marks are typically performed one at a time in a population of cells, and their colocalization is inferred by association. Here, we describe a single-molecule analytical approach that can perform direct detection of multiple epigenetic marks simultaneously and use it to identify mechanisms coordinating placement of three gene silencing marks, trimethylated histone H3 lysine 9, lysine 27 (H3K9me3, H3K27me3), and cytosine methylation (mC), in the normal and cancer genome. We show that H3K9me3 and mC are present together on individual chromatin fragments in mouse embryonic stem cells and that half of the H3K9me3 marks require mC for their placement. In contrast, mC and H3K27me3 coincidence is rare, and in fact, mC antagonizes H3K27me3 in both embryonic stem cells and primary mouse fibroblasts, indicating this antagonism is shared among primary cells. However, upon immortalization or tumorigenic transformation of mouse fibroblasts, mC is required for complete H3K27me3 placement. Importantly, in human promyelocytic cells, H3K27me3 is also dependent on mC. Because aberrant placement of gene silencing marks at tumor suppressor genes contributes to tumor progression, the improper dependency of H3K27me3 by mC in immortalized cells is likely to be fundamental to cancer. Our platform can enable other studies involving coordination of epigenetic marks and leverage efforts to discover disease biomarkers and epigenome-modifying drugs.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/chemistry , Animals , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Cytosine/chemistry , Epigenomics , Fibroblasts/metabolism , Gene Silencing , Humans , Lysine/genetics , Methylation , Mice , Protein Binding
4.
Mol Reprod Dev ; 82(12): 932-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26517592

ABSTRACT

Noncoding RNAs (ncRNAs) have long been known to play vital roles in eukaryotic gene regulation. Studies conducted over a decade ago revealed that maturation of spliced, polyadenylated coding mRNA occurs by reactions involving small nuclear RNAs and small nucleolar RNAs; mRNA translation depends on activities mediated by transfer RNAs and ribosomal RNAs, subject to negative regulation by micro RNAs; transcriptional competence of sex chromosomes and some imprinted genes is regulated in cis by ncRNAs that vary by species; and both small-interfering RNAs and piwi-interacting RNAs bound to Argonaute-family proteins regulate post-translational modifications on chromatin and local gene expression states. More recently, gene-regulating noncoding RNAs have been identified, such as long intergenic and long noncoding RNAs (collectively referred to as lncRNAs)--a class totaling more than 100,000 transcripts in humans, which include some of the previously mentioned RNAs that regulate dosage compensation and imprinted gene expression. Here, we provide an overview of lncRNA activities, and then review the role of lncRNAs in processes vital to reproduction, such as germ cell specification, sex determination and gonadogenesis, sex hormone responses, meiosis, gametogenesis, placentation, non-genetic inheritance, and pathologies affecting reproductive tissues. Results from many species are presented to illustrate the evolutionarily conserved processes lncRNAs are involved in.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , Gene Expression Regulation, Developmental/physiology , Genomic Imprinting/physiology , RNA, Long Noncoding/metabolism , Reproduction/physiology , Animals , Chromatin/genetics , Humans , RNA, Long Noncoding/genetics
5.
Proc Natl Acad Sci U S A ; 109(22): 8477-82, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586076

ABSTRACT

Epigenetic modifications, such as DNA and histone methylation, are responsible for regulatory pathways that affect disease. Current epigenetic analyses use bisulfite conversion to identify DNA methylation and chromatin immunoprecipitation to collect molecules bearing a specific histone modification. In this work, we present a proof-of-principle demonstration for a new method using a nanofluidic device that combines real-time detection and automated sorting of individual molecules based on their epigenetic state. This device evaluates the fluorescence from labeled epigenetic modifications to actuate sorting. This technology has demonstrated up to 98% accuracy in molecule sorting and has achieved postsorting sample recovery on femtogram quantities of genetic material. We have applied it to sort methylated DNA molecules using simultaneous, multicolor fluorescence to identify methyl binding domain protein-1 (MBD1) bound to full-duplex DNA. The functionality enabled by this nanofluidic platform now provides a workflow for color-multiplexed detection, sorting, and recovery of single molecules toward subsequent DNA sequencing.


Subject(s)
DNA Methylation , DNA/genetics , Microfluidic Analytical Techniques/methods , Nanotechnology/methods , DNA/analysis , DNA/metabolism , DNA-Binding Proteins/metabolism , Fluorescence , Humans , Microfluidic Analytical Techniques/instrumentation , Microscopy, Confocal , Nanotechnology/instrumentation , Protein Binding , Real-Time Polymerase Chain Reaction/methods , Reproducibility of Results , Time Factors , Transcription Factors/metabolism
6.
Nat Genet ; 32(3): 426-31, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12410230

ABSTRACT

Genomic imprinting is an epigenetic modification that results in expression from only one of the two parental copies of a gene. Differences in methylation between the two parental chromosomes are often observed at or near imprinted genes. Beckwith-Wiedemann syndrome (BWS), which predisposes to cancer and excessive growth, results from a disruption of imprinted gene expression in chromosome band 11p15.5. One third of individuals with BWS lose maternal-specific methylation at KvDMR1, a putative imprinting control region within intron 10 of the KCNQ1 gene, and it has been proposed that this epimutation results in aberrant imprinting and, consequently, BWS1, 2. Here we show that paternal inheritance of a deletion of KvDMR1 results in the de-repression in cis of six genes, including Cdkn1c, which encodes cyclin-dependent kinase inhibitor 1C. Furthermore, fetuses and adult mice that inherited the deletion from their fathers were 20-25% smaller than their wildtype littermates. By contrast, maternal inheritance of this deletion had no effect on imprinted gene expression or growth. Thus, the unmethylated paternal KvDMR1 allele regulates imprinted expression by silencing genes on the paternal chromosome. These findings support the hypothesis that loss of methylation in BWS patients activates the repressive function of KvDMR1 on the maternal chromosome, resulting in abnormal silencing of CDKN1C and the development of BWS.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , Brain/metabolism , Genomic Imprinting , Oligonucleotide Array Sequence Analysis , Alleles , Animals , Blotting, Northern , Chromosome Mapping , Down-Regulation , Fathers , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Molecular Sequence Data , Mothers , Physical Chromosome Mapping , Up-Regulation
7.
Nat Genet ; 34(2): 199-202, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12740578

ABSTRACT

In mammals, imprinted genes have parent-of-origin-specific patterns of DNA methylation that cause allele-specific expression. At Rasgrf1 (encoding RAS protein-specific guanine nucleotide-releasing factor 1), a repeated DNA element is needed to establish methylation and expression of the active paternal allele. At Igf2r (encoding insulin-like growth factor 2 receptor), a sequence called region 2 is needed for methylation of the active maternal allele. Here we show that replacing the Rasgrf1 repeats on the paternal allele with region 2 allows both methylation and expression of the paternal copy of Rasgrf1, indicating that sequences that control methylation can function ectopically. Paternal transmission of the mutated allele also induced methylation and expression in trans of the normally unmethylated and silent wild-type maternal allele. Once activated, the wild-type maternal Rasgrf1 allele maintained its activated state in the next generation independently of the paternal allele. These results recapitulate in mice several features in common with paramutation described in plants.


Subject(s)
DNA Methylation , Mutation , Alleles , Animals , Crosses, Genetic , DNA-Binding Proteins , Female , GTP-Binding Proteins/genetics , GTPase-Activating Proteins , Gene Silencing , Genomic Imprinting , Male , Mice , Mice, Mutant Strains , Repetitive Sequences, Nucleic Acid , Repressor Proteins , ras-GRF1
8.
Nat Genet ; 30(1): 92-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11753386

ABSTRACT

In mammals, DNA is methylated at cytosines within CpG dinucleotides. Properly regulated methylation is crucial for normal development. Inappropriate methylation may contribute to tumorigenesis by silencing tumor-suppressor genes or by activating growth-stimulating genes. Although many genes have been identified that acquire methylation and whose expression is methylation-sensitive, little is known about how DNA methylation is controlled. We have identified a DNA sequence that regulates establishment of DNA methylation in the male germ line at Rasgrf1. In mice, the imprinted Rasgrf1 locus is methylated on the paternal allele within a differentially methylated domain (DMD) 30 kbp 5' of the promoter. Expression is exclusively from the paternal allele in neonatal brain. Methylation is regulated by a repeated sequence, consisting of a 41-mer repeated 40 times, found immediately 3' of the DMD. This sequence is present in organisms in which Rasgrf1 is imprinted. In addition, DMD methylation is required for imprinted Rasgrf1 expression. Together the DMD and repeat element constitute a binary switch that regulates imprinting at the locus.


Subject(s)
DNA Methylation , ras-GRF1/genetics , Alleles , Animals , CpG Islands , Crosses, Genetic , Female , Gene Expression Regulation , Gene Targeting , Genomic Imprinting , Male , Mice , Mice, Inbred C57BL , Polymorphism, Restriction Fragment Length , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
9.
Pigment Cell Melanoma Res ; 36(6): 531-541, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37462349

ABSTRACT

Melanocyte stem cells (McSCs) of the hair follicle are necessary for hair pigmentation and can serve as melanoma cells of origin when harboring cancer-driving mutations. McSCs can be released from quiescence, activated, and undergo differentiation into pigment-producing melanocytes during the hair cycle or due to environmental stimuli, such as ultraviolet-B (UVB) exposure. However, our current understanding of the mechanisms regulating McSC stemness, activation, and differentiation remains limited. Here, to capture the differing possible states in which murine McSCs can exist, we sorted melanocyte nuclei from quiescent (telogen) skin, skin actively producing hair shafts (anagen), and skin exposed to UVB. With these sorted nuclei, we then utilized single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing (snATAC-seq) and characterized three melanocyte lineages: quiescent McSCs (qMcSCs), activated McSCs (aMcSCs), and differentiated melanocytes (dMCs) that co-exist in all three skin conditions. Furthermore, we successfully identified differentially accessible genes and enriched transcription factor binding motifs for each melanocyte lineage. Our findings reveal potential gene regulators that determine these melanocyte cell states and provide new insights into how aMcSC chromatin states are regulated differently under divergent intrinsic and extrinsic cues. We also provide a publicly available online tool with a user-friendly interface to explore this comprehensive dataset, which will provide a resource for further studies on McSC regulation upon natural or UVB-mediated stem cell activation.


Subject(s)
Chromatin , Melanocytes , Mice , Animals , Chromatin/metabolism , Melanocytes/metabolism , Skin , Hair Follicle/metabolism , Stem Cells , Cell Differentiation
10.
Nutrients ; 15(16)2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37630723

ABSTRACT

Non-shivering thermogenesis (NST) has strong potential to combat obesity; however, a safe molecular approach to activate this process has not yet been identified. The sulfur amino acid taurine has the ability to safely activate NST and confer protection against obesity and metabolic disease in both mice and humans, but the mechanism of this action is unknown. In this study, we discover that a suite of taurine biosynthetic enzymes, especially that of cysteamine dioxygenase (ADO), significantly increases in response to ß3 adrenergic signaling in inguinal adipose tissue (IWAT) in order to increase intracellular concentrations of taurine. We further show that ADO is critical for thermogenic mitochondrial respiratory function as its ablation in adipocytes significantly reduces taurine levels, which leads to declines in mitochondrial oxygen consumption rates. Finally, we demonstrate via assay for transposase-accessible chromatin with sequencing (ATAC-seq) that taurine supplementation in beige adipocytes has the ability to remodel the chromatin landscape to increase the chromatin accessibility and transcription of genes, such as glucose-6-phosphate isomerase 1 (Gpi1), which are critical for NST. Taken together, our studies highlight a potential mechanism for taurine in the activation of NST that can be leveraged toward the treatment of obesity and metabolic disease.


Subject(s)
Adipose Tissue , Chromatin , Humans , Animals , Mice , Respiratory Rate , Adipocytes , Respiration
11.
Circ Res ; 106(4): 796-808, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20056917

ABSTRACT

RATIONALE: Myocardial infarction (MI) results in remodeling of the myocardium and the extracellular matrix (ECM). Tissue inhibitors of metalloproteinases (TIMPs) are critical regulators of ECM integrity via inhibiting matrix metalloproteinases (MMPs). TIMP2 is highly expressed in the heart and is the only TIMP that, in addition to inhibiting MMPs, is required for cell surface activation of pro-MMP2. Hence, it is difficult to predict the function of TIMP2 as protective (MMP-inhibiting) or harmful (MMP-activating) in heart disease. OBJECTIVE: We examined the role of TIMP2 in the cardiac response to MI. METHODS AND RESULTS: MI was induced in 11- to 12-week-old male TIMP2(-/-) and age-matched wild-type mice. Cardiac function was monitored by echocardiography at 1 and 4 weeks post-MI. ECM fibrillar structure was visualized using second harmonic generation and multiphoton imaging of unfixed/unstained hearts. Molecular analyses were performed at 3 days and 1 week post-MI on flash-frozen infarct, periinfarct, and noninfarct tissue. Membrane type 1 (MT1)-MMP levels and activity were measured in membrane protein fractions. TIMP2(-/-)-MI mice exhibited a 25% greater infarct expansion, markedly exacerbated left ventricular dilation (by 12%) and dysfunction (by 30%), and more severe inflammation compared to wild-type MI mice. Adverse ECM remodeling was detected by reduced density and enhanced disarray of fibrillar collagen in TIMP2(-/-)-MI compared to wild-type MI hearts. TIMP2 deficiency completely abrogated MMP2 activation but markedly increased collagenase activity, particularly MT1-MMP activity post-MI. CONCLUSIONS: The MMP-inhibitory function of TIMP2 is a key determinant of post-MI myocardial remodeling primarily because of its inhibitory action on MT1-MMP. TIMP2 replenishment in diseased myocardium could provide a potential therapy in reducing or preventing disease progression.


Subject(s)
Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Myocardial Infarction/enzymology , Myocardium/enzymology , Tissue Inhibitor of Metalloproteinase-2/deficiency , Ventricular Remodeling , Animals , Disease Models, Animal , Echocardiography, Doppler , Enzyme Activation , Extracellular Matrix/metabolism , Fibrillar Collagens/metabolism , Inflammation/enzymology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence, Multiphoton , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardium/pathology , RNA, Messenger/metabolism , Recovery of Function , Severity of Illness Index , Stroke Volume , Time Factors , Tissue Inhibitor of Metalloproteinase-2/genetics , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
12.
Commun Biol ; 5(1): 584, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35701601

ABSTRACT

Beige adipocytes are induced by cold temperatures or ß3-adrenergic receptor (Adrb3) agonists. They create heat through glucose and fatty acid (FA) oxidation, conferring metabolic benefits. The distinct and shared mechanisms by which these treatments induce beiging are unknown. Here, we perform single-nucleus assay for transposase-accessible chromatin sequencing (snATAC-seq) on adipose tissue from mice exposed to cold or an Adrb3 agonist to identify cellular and chromatin accessibility dynamics during beiging. Both stimuli induce chromatin remodeling that influence vascularization and inflammation in adipose. Beige adipocytes from cold-exposed mice have increased accessibility at genes regulating glycolytic processes, whereas Adrb3 activation increases cAMP responses. While both thermogenic stimuli increase accessibility at genes regulating thermogenesis, lipogenesis, and beige adipocyte development, the kinetics and magnitudes of the changes are distinct for the stimuli. Accessibility changes at lipogenic genes are linked to functional changes in lipid composition of adipose. Both stimuli tend to decrease the proportion of palmitic acids, a saturated FA in adipose. However, Adrb3 activation increases the proportion of monounsaturated FAs, whereas cold increases the proportion of polyunsaturated FAs. These findings reveal common and distinct mechanisms of cold and Adrb3 induced beige adipocyte biogenesis, and identify unique functional consequences of manipulating these pathways in vivo.


Subject(s)
Adipocytes, Beige , Gene Regulatory Networks , Adipocytes, Beige/metabolism , Adipose Tissue , Animals , Chromatin/metabolism , Mice , Thermogenesis/genetics
13.
Cancer Res Commun ; 2(7): 663-678, 2022 07.
Article in English | MEDLINE | ID: mdl-36923282

ABSTRACT

Fibrolamellar carcinoma (FLC) is an aggressive liver cancer with no effective therapeutic options. The extracellular environment of FLC tumors is poorly characterized and may contribute to cancer growth and/or metastasis. To bridge this knowledge gap, we assessed pathways relevant to proteoglycans, a major component of the extracellular matrix. We first analyzed gene expression data from FLC and nonmalignant liver tissue (n = 27) to identify changes in glycosaminoglycan (GAG) biosynthesis pathways and found that genes associated with production of chondroitin sulfate, but not other GAGs, are significantly increased by 8-fold. We then implemented a novel LC/MS-MS based method to quantify the abundance of different types of GAGs in patient tumors (n = 16) and found that chondroitin sulfate is significantly more abundant in FLC tumors by 6-fold. Upon further analysis of GAG-associated proteins, we found that versican (VCAN) expression is significantly upregulated at the mRNA and protein levels, the latter of which was validated by IHC. Finally, we performed single-cell assay for transposase-accessible chromatin sequencing on FLC tumors (n = 3), which revealed for the first time the different cell types in FLC tumors and also showed that VCAN is likely produced not only from FLC tumor epithelial cells but also activated stellate cells. Our results reveal a pathologic aberrancy in chondroitin (but not heparan) sulfate proteoglycans in FLC and highlight a potential role for activated stellate cells. Significance: This study leverages a multi-disciplinary approach, including state-of-the-art chemical analyses and cutting-edge single-cell genomic technologies, to identify for the first time a marked chondroitin sulfate aberrancy in FLC that could open novel therapeutic avenues in the future.


Subject(s)
Carcinoma, Hepatocellular , Chondroitin Sulfates , Humans , Chondroitin Sulfates/metabolism , Carcinoma, Hepatocellular/genetics , Heparan Sulfate Proteoglycans , Versicans
14.
PLoS Genet ; 4(8): e1000145, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18670629

ABSTRACT

At the imprinted Rasgrf1 locus in mouse, a cis-acting sequence controls DNA methylation at a differentially methylated domain (DMD). While characterizing epigenetic marks over the DMD, we observed that DNA and H3K27 trimethylation are mutually exclusive, with DNA and H3K27 methylation limited to the paternal and maternal sequences, respectively. The mutual exclusion arises because one mark prevents placement of the other. We demonstrated this in five ways: using 5-azacytidine treatments and mutations at the endogenous locus that disrupt DNA methylation; using a transgenic model in which the maternal DMD inappropriately acquired DNA methylation; and by analyzing materials from cells and embryos lacking SUZ12 and YY1. SUZ12 is part of the PRC2 complex, which is needed for placing H3K27me3, and YY1 recruits PRC2 to sites of action. Results from each experimental system consistently demonstrated antagonism between H3K27me3 and DNA methylation. When DNA methylation was lost, H3K27me3 encroached into sites where it had not been before; inappropriate acquisition of DNA methylation excluded normal placement of H3K27me3, and loss of factors needed for H3K27 methylation enabled DNA methylation to appear where it had been excluded. These data reveal the previously unknown antagonism between H3K27 and DNA methylation and identify a means by which epigenetic states may change during disease and development.


Subject(s)
DNA Methylation , Genomic Imprinting , Histones/metabolism , Lysine/metabolism , ras-GRF1/genetics , Alleles , Animals , Cells, Cultured , Female , Male , Methylation , Mice , Mice, Inbred Strains , Models, Genetic , Species Specificity , ras-GRF1/metabolism
15.
PLoS One ; 16(12): e0261783, 2021.
Article in English | MEDLINE | ID: mdl-34972124

ABSTRACT

Obesity promotes type 2 diabetes and cardiometabolic pathologies. Vertical sleeve gastrectomy (VSG) is used to treat obesity resulting in long-term weight loss and health improvements that precede weight loss; however, the mechanisms underlying the immediate benefits remain incompletely understood. Because adipose plays a crucial role in energy homeostasis and utilization, we hypothesized that VSG exerts its influences, in part, by modulating adipose functional states. We applied single-cell ATAC sequencing and lipid profiling to inguinal and epididymal adipose depots from mice that received sham surgery or VSG. We observed depot-specific cellular composition and chromatin accessibility patterns that were altered by VSG. Specifically, accessibility at Scd1, a fatty acid desaturase, was substantially reduced after VSG in mature adipocytes of inguinal but not epididymal depots. This was accompanied by reduced accumulation of SCD1-produced unsaturated fatty acids. Given these findings and reports that reductions in Scd1 attenuate obesity and insulin resistance our results suggest VSG exerts its beneficial effects through an inguinal depot-specific reduction of SCD1 activity.


Subject(s)
Chromatin , Diabetes Mellitus, Type 2 , Animals , Bariatric Surgery , Gastrectomy , Mice , Weight Loss
16.
Anal Chem ; 82(6): 2480-7, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20184350

ABSTRACT

Epigenetic states are governed by DNA methylation and a host of modifications to histones bound with DNA. These states are essential for proper developmentally regulated gene expression and are perturbed in many diseases. There is great interest in identifying epigenetic mark placement genome wide and understanding how these marks vary among cell types, with changes in environment or according to health and disease status. Current epigenomic analyses employ bisulfite sequencing and chromatin immunoprecipitation, but query only one type of epigenetic mark at a time, DNA methylation, or histone modifications and often require substantial input material. To overcome these limitations, we established a method using nanofluidics and multicolor fluorescence microscopy to detect DNA and histones in individual chromatin fragments at about 10 Mbp/min. We demonstrated its utility for epigenetic analysis by identifying DNA methylation on individual molecules. This technique will provide the unprecedented opportunity for genome wide, simultaneous analysis of multiple epigenetic states on single molecules.


Subject(s)
Chromatin/chemistry , DNA Methylation , DNA/analysis , Histones/chemistry , Microfluidics/instrumentation , Microscopy, Fluorescence/methods , Epigenesis, Genetic , Equipment Design , HeLa Cells , Humans
17.
Mamm Genome ; 20(9-10): 654-63, 2009.
Article in English | MEDLINE | ID: mdl-19513790

ABSTRACT

Rasgrf1 is imprinted and expressed preferentially from the paternal allele in neonatal mouse brain. At weaning, expression becomes biallelic. Using a mouse model, we assayed the effects of perturbing imprinted Rasgrf1 expression in mice with the following imprinted expression patterns: monoallelic paternal (wild type), monoallelic maternal (maternal only), biallelic (both alleles transcribed), and null (neither allele transcribed). All genotypes exhibit biallelic expression around weaning. Consequences of this transient imprinting perturbation are manifested as overall size differences that correspond to the amount of neonatal Rasgrf1 expressed and are persistent, extending into adulthood. Biallelic mice are the largest and overexpress Rasgrf1 relative to wild-type mice, null mice are the smallest and underexpress Rasgrf1 as neonates, and the two monoallelically expressing genotypes are intermediate and indistinguishable from one another, in both size and Rasgrf1 expression level. Importantly, these data support one of the key underlying assumptions of the "conflict hypothesis" that describes the evolution of genomic imprinting in mammals and supposes that equivalent amounts of imprinted gene expression produce equivalent phenotypes, regardless of which parental allele is transcribed. Concordant with the difference in overall body size, we identify differences in IGF-1 levels, both in serum protein and as liver transcript, and identify additional differential expression of components upstream of IGF-1 release in the GH/IGF-1 axis. These data suggest that imprinted Rasgrf1 expression affects GH/IGF-1 axis function, and that the consequences of Rasgrf1 inputs to this axis persist beyond the time period when expression is restricted via epigenetic mechanisms, suggesting that proper neonatal Rasgrf1 expression levels are critical for development.


Subject(s)
Genomic Imprinting , Insulin-Like Growth Factor I/metabolism , Mice/growth & development , Mutation , ras-GRF1/genetics , Animals , Body Size , Female , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor I/genetics , Male , Mice/genetics , Mice/metabolism , Mice, Inbred C57BL , ras-GRF1/metabolism
18.
Mol Cell Biol ; 26(24): 9564-70, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17030618

ABSTRACT

Epigenetic programming is critical for normal development of mammalian embryos. Errors cause misexpression of genes and aberrant development (E. Li, C. Beard, and R. Jaenisch, Nature 366:362-365, 1993). Imprinted genes are important targets of epigenetic regulation, but little is known about how the epigenetic patterns are established in the parental germ lines and maintained in the embryo. Paternal allele-specific expression at the imprinted Rasgrf1 locus in mice is controlled by paternal allele-specific methylation at a differentially methylated domain (DMD). DMD methylation is in turn controlled by a direct repeat sequence immediately downstream of the DMD which is required for establishing Rasgrf1 methylation in the male germ line (B. J. Yoon et al., Nat. Genet. 30:92-96, 2002). To determine if these repeats have a role in methylation maintenance, we developed a conditional deletion of the repeat sequence in mice and showed that the repeats are also required during a narrow interval to maintain paternal methylation of Rasgrf1 in developing embryos. Removing the repeats upon fertilization caused a total loss of methylation by the morula stage, but by the epiblast stage, the repeats were completely dispensable for methylation maintenance. This developmental interval coincides with genome-wide demethylation and remethylation in mice which most imprinted genes resist. Our data show that the Rasgrf1 repeats serve at least two functions: first, to establish Rasgrf1 DNA methylation in the male germ line, and second, to resist global demethylation in the preimplantation embryo.


Subject(s)
DNA Methylation , Gene Expression Regulation, Developmental/physiology , Genomic Imprinting/physiology , Repetitive Sequences, Nucleic Acid , ras-GRF1/genetics , Animals , Blastocyst/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Mol Cell Biol ; 25(24): 11184-90, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314537

ABSTRACT

Imprinted methylation of the paternal Rasgrf1 allele in mice occurs at a differentially methylated domain (DMD) 30 kbp 5' of the promoter. A repeated sequence 3' of the DMD regulates imprinted methylation, which is required for imprinted expression. Here we identify the mechanism by which methylation controls imprinting. The DMD is an enhancer blocker that binds CTCF in a methylation-sensitive manner. CTCF bound to the unmethylated maternal allele silences expression. CTCF binding to the paternal allele is prevented by repeat-mediated methylation, allowing expression. Optimal in vitro enhancer-blocking activity requires CTCF binding sites. The enhancer blocker can be bypassed in vivo and imprinting abolished by placing an extra enhancer proximal to the promoter. Together, the repeats and the DMD constitute a binary switch that regulates Rasgrf1 imprinting.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Genomic Imprinting , Repressor Proteins/metabolism , Animals , CCCTC-Binding Factor , GTPase-Activating Proteins , Mice , Models, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL