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










Publication year range
1.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716726

ABSTRACT

Childhood-onset essential hypertension (COEH) is an uncommon form of hypertension that manifests in childhood or adolescence and, in the United States, disproportionately affects children of African ancestry. The etiology of COEH is unknown, but its childhood onset, low prevalence, high heritability, and skewed ancestral demography suggest the potential to identify rare genetic variation segregating in a Mendelian manner among affected individuals and thereby implicate genes important to disease pathogenesis. However, no COEH genes have been reported to date. Here, we identify recessive segregation of rare and putatively damaging missense variation in the spectrin domain of spectrin repeat containing nuclear envelope protein 1 (SYNE1), a cardiovascular candidate gene, in 3 of 16 families with early-onset COEH without an antecedent family history. By leveraging exome sequence data from an additional 48 COEH families, 1,700 in-house trios, and publicly available data sets, we demonstrate that compound heterozygous SYNE1 variation in these COEH individuals occurred more often than expected by chance and that this class of biallelic rare variation was significantly enriched among individuals of African genetic ancestry. Using in vitro shRNA knockdown of SYNE1, we show that reduced SYNE1 expression resulted in a substantial decrease in the elasticity of smooth muscle vascular cells that could be rescued by pharmacological inhibition of the downstream RhoA/Rho-associated protein kinase pathway. These results provide insights into the molecular genetics and underlying pathophysiology of COEH and suggest a role for precision therapeutics in the future.


Subject(s)
Cytoskeletal Proteins , Essential Hypertension , Exome Sequencing , Nerve Tissue Proteins , Adolescent , Child , Female , Humans , Male , Age of Onset , Cytoskeletal Proteins/genetics , Essential Hypertension/genetics , Exome/genetics , Genetic Predisposition to Disease , Mutation, Missense/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Pedigree , rhoA GTP-Binding Protein/genetics , United States/epidemiology , Infant, Newborn , Infant , Child, Preschool , Young Adult
2.
Proc Natl Acad Sci U S A ; 115(29): 7611-7616, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967166

ABSTRACT

Brain "inflammaging," a low-grade and chronic inflammation, is a major hallmark for aging-related neurodegenerative diseases. Here, by profiling H3K27ac and gene expression patterns in human and mouse brains, we found that age-related up-regulated (Age-Up) and down-regulated (Age-Down) genes have distinct H3K27ac patterns. Although both groups show promoter H3K27ac, the Age-Up genes, enriched for inflammation-related functions, are additionally marked by broad H3K27ac distribution over their gene bodies, which is progressively reduced during aging. Age-related gene expression changes can be predicted by gene-body H3K27ac level. Contrary to the presumed transcription activation function of promoter H3K27ac, we found that broad gene-body hyper H3K27ac suppresses overexpression of inflammaging genes. Altogether, our findings revealed opposite regulations by H3K27ac of Age-Up and Age-Down genes and a mode of broad gene-body H3K27ac in repressing transcription.


Subject(s)
Aging/metabolism , Brain/metabolism , Histones/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Transcriptome , Acetylation , Aging/genetics , Animals , Gene Expression Profiling , Histones/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Mice
3.
Sci Rep ; 7(1): 1136, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28442710

ABSTRACT

Epigenetic mechanisms are known to exert control over gene expression and determine cell fate. Genetic mutations in epigenetic regulators are responsible for several neurologic disorders. Mutations of the chromatin remodeling protein Lsh/HELLS can cause the human Immunodeficiency, Centromere instability and Facial anomalies (ICF) syndrome, which is associated with neurologic deficiencies. We report here a critical role for Lsh in murine neural development. Lsh depleted neural stem/progenitor cells (NSPCs) display reduced growth, increases in apoptosis and impaired ability of self-renewal. RNA-seq analysis demonstrates differential gene expression in Lsh-/- NSPCs and suggests multiple aberrant pathways. Concentrating on specific genomic targets, we show that ablation of Lsh alters epigenetic states at specific enhancer regions of the key cell cycle regulator Cdkn1a and the stem cell regulator Bmp4 in NSPCs and alters their expression. These results suggest that Lsh exerts epigenetic regulation at key regulators of neural stem cell fate ensuring adequate NSPCs self-renewal and maintenance during development.


Subject(s)
Cell Self Renewal , DNA Helicases/metabolism , Epigenesis, Genetic , Neural Stem Cells/physiology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Gene Knockout Techniques , Mice , Sequence Analysis, RNA
4.
Bioinform Biol Insights ; 10: 267-289, 2016.
Article in English | MEDLINE | ID: mdl-27980397

ABSTRACT

Epigenetics is one of the most rapidly expanding fields in biomedical research, and the popularity of the high-throughput next-generation sequencing (NGS) highlights the accelerating speed of epigenomics discovery over the past decade. Epigenetics studies the heritable phenotypes resulting from chromatin changes but without alteration on DNA sequence. Epigenetic factors and their interactive network regulate almost all of the fundamental biological procedures, and incorrect epigenetic information may lead to complex diseases. A comprehensive understanding of epigenetic mechanisms, their interactions, and alterations in health and diseases genome widely has become a priority in biological research. Bioinformatics is expected to make a remarkable contribution for this purpose, especially in processing and interpreting the large-scale NGS datasets. In this review, we introduce the epigenetics pioneering achievements in health status and complex diseases; next, we give a systematic review of the epigenomics data generation, summarize public resources and integrative analysis approaches, and finally outline the challenges and future directions in computational epigenomics.

5.
Bioinform Biol Insights ; 9(Suppl 1): 29-46, 2015.
Article in English | MEDLINE | ID: mdl-26609224

ABSTRACT

Next-generation sequencing technologies have revolutionarily advanced sequence-based research with the advantages of high-throughput, high-sensitivity, and high-speed. RNA-seq is now being used widely for uncovering multiple facets of transcriptome to facilitate the biological applications. However, the large-scale data analyses associated with RNA-seq harbors challenges. In this study, we present a detailed overview of the applications of this technology and the challenges that need to be addressed, including data preprocessing, differential gene expression analysis, alternative splicing analysis, variants detection and allele-specific expression, pathway analysis, co-expression network analysis, and applications combining various experimental procedures beyond the achievements that have been made. Specifically, we discuss essential principles of computational methods that are required to meet the key challenges of the RNA-seq data analyses, development of various bioinformatics tools, challenges associated with the RNA-seq applications, and examples that represent the advances made so far in the characterization of the transcriptome.

6.
Epigenetics ; 10(9): 861-71, 2015.
Article in English | MEDLINE | ID: mdl-26252449

ABSTRACT

Several studies have described phenotypic changes in the offspring of mice exposed to a variety of environmental factors, including diet, toxins, and stress; however, the molecular pathways involved in these changes remain unclear. Using a high fat diet (HFD)-induced obesity mouse model, we examined liver gene expression in male offspring and analyzed chromatin of paternal spermatozoa. We found that the hepatic mRNA level of 7 genes (out of 20 evaluated) was significantly altered in HFD male offspring compared to control mice, suggesting that phenotypic changes in the offspring depend on parental diet. We examined 7 imprinted loci in spermatozoa DNA from HFD-treated and control fathers by bisulfite sequencing, but did not detect changes in DNA methylation associated with HFD. Using chromatin immunoprecipitation followed by high-throughput sequencing, we found differential histone H3-occupancy at genes involved in the regulation of embryogenesis and differential H3K4me1-enrichment at transcription regulatory genes in HFD fathers vs. control mice. These results suggest that dietary exposure can modulate histone composition at regulatory genes implicated in developmental processes.


Subject(s)
Diet, High-Fat/adverse effects , Gene Expression/drug effects , Histones/metabolism , Liver/metabolism , Spermatozoa/drug effects , Animals , DNA Methylation/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Genomic Imprinting/drug effects , Male , Mice , Sequence Analysis, DNA , Spermatozoa/metabolism
7.
Nucleic Acids Res ; 43(3): 1444-55, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25578963

ABSTRACT

Lsh, a chromatin remodeling protein of the SNF2 family, is critical for normal heterochromatin structure. In particular, DNA methylation at repeat elements, a hallmark of heterochromatin, is greatly reduced in Lsh(-/-) (KO) cells. Here, we examined the presumed nucleosome remodeling activity of Lsh on chromatin in the context of DNA methylation. We found that dynamic CG methylation was dependent on Lsh in embryonic stem cells. Moreover, we demonstrate that ATP function is critical for de novo methylation at repeat sequences. The ATP binding site of Lsh is in part required to promote stable association of the DNA methyltransferase 3b with the repeat locus. By performing nucleosome occupancy assays, we found distinct nucleosome occupancy in KO ES cells compared to WT ES cells after differentiation. Nucleosome density was restored to wild-type level by re-expressing wild-type Lsh but not the ATP mutant in KO ES cells. Our results suggest that ATP-dependent nucleosome remodeling is the primary molecular function of Lsh, which may promote de novo methylation in differentiating ES cells.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA Methylation , Nucleosomes/metabolism , Repetitive Sequences, Nucleic Acid , Binding Sites , Cells, Cultured , Humans
8.
Genome Res ; 24(10): 1613-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25170028

ABSTRACT

Cytosine methylation is critical in mammalian development and plays a role in diverse biologic processes such as genomic imprinting, X chromosome inactivation, and silencing of repeat elements. Several factors regulate DNA methylation in early embryogenesis, but their precise role in the establishment of DNA methylation at a given site remains unclear. We have generated a comprehensive methylation map in fibroblasts derived from the murine DNA methylation mutant Hells(-/-) (helicase, lymphoid specific, also known as LSH). It has been previously shown that HELLS can influence de novo methylation of retroviral sequences and endogenous genes. Here, we describe that HELLS controls cytosine methylation in a nuclear compartment that is in part defined by lamin B1 attachment regions. Despite widespread loss of cytosine methylation at regulatory sequences, including promoter regions of protein-coding genes and noncoding RNA genes, overall relative transcript abundance levels in the absence of HELLS are similar to those in wild-type cells. A subset of promoter regions shows increases of the histone modification H3K27me3, suggesting redundancy of epigenetic silencing mechanisms. Furthermore, HELLS modulates CG methylation at all classes of repeat elements and is critical for repression of a subset of repeat elements. Overall, we provide a detailed analysis of gene expression changes in relation to DNA methylation alterations, which contributes to our understanding of the biological role of cytosine methylation.


Subject(s)
Cytosine/metabolism , DNA Helicases/genetics , DNA Methylation , DNA/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Nucleus/genetics , Embryonic Stem Cells , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockout Techniques , Histones/metabolism , Lamin Type B/metabolism , Mice , Molecular Sequence Data , Mutation , Regulatory Sequences, Nucleic Acid , Repetitive Sequences, Nucleic Acid
9.
Proc Natl Acad Sci U S A ; 111(16): 5890-5, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711395

ABSTRACT

DNA methylation patterns are established in early embryogenesis and are critical for cellular differentiation. To investigate the role of CG methylation in potential enhancer formation, we assessed H3K4me1 modification in murine embryonic fibroblasts (MEFs) derived from the DNA methylation mutant Lsh(-/-) mice. We report here de novo formation of putative enhancer elements at CG hypomethylated sites that can be dynamically altered. We found a subset of differentially enriched H3K4me1 regions clustered at neuronal lineage genes and overlapping with known cis-regulatory elements present in brain tissue. Reprogramming of Lsh(-/-) MEFs into induced pluripotent stem (iPS) cells leads to increased neuronal lineage gene expression of premarked genes and enhanced differentiation potential of Lsh(-/-) iPS cells toward the neuronal lineage pathway compared with WT iPS cells in vitro and in vivo. The state of CG hypomethylation and H3K4me1 enrichment is partially maintained in Lsh(-/-) iPS cells. The acquisition of H3K27ac and activity of subcloned fragments in an enhancer reporter assay indicate functional activity of several of de novo H3K4me1-marked sequences. Our results suggest a functional link of H3K4me1 enrichment at CG hypomethylated sites, enhancer formation, and cellular plasticity.


Subject(s)
CpG Islands/genetics , DNA Helicases/deficiency , DNA Methylation/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Biomarkers/metabolism , Cell Lineage , DNA Helicases/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Fibroblasts/cytology , HeLa Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Neurons/cytology , Protein Binding , Signal Transduction , Transcription Factors/metabolism
10.
Aging Cell ; 11(6): 1055-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22978322

ABSTRACT

Epigenetic modifications are critical determinants of cellular and developmental states. Epigenetic changes, such as decreased H3K27me3 histone methylation on insulin/IGF1 genes, have been previously shown to modulate lifespan through gene expression regulation. However, global epigenetic changes during aging and their biological functions, if any, remain elusive. Here, we examined the histone modification H3K4 dimethylation (H3K4me2) in the prefrontal cortex of individual rhesus macaques at different ages by chromatin immunoprecipitation, followed by deep sequencing (ChIP-seq) at the whole genome level. Through integrative analysis of the ChIP-seq profiles with gene expression data, we found that H3K4me2 increased at promoters and enhancers globally during postnatal development and aging, and those that correspond to gene expression changes in cis are enriched for stress responses, such as the DNA damage response. This suggests that metabolic and environmental stresses experienced by an organism are associated with the progressive opening of chromatin. In support of this, we also observed increased expression of two H3K4 methyltransferases, SETD7 and DPY30, in aged macaque brain.


Subject(s)
Aging/genetics , Brain/metabolism , Chromatin/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genome , Histones/metabolism , Aging/metabolism , Animals , Brain/growth & development , Chromatin/metabolism , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Macaca mulatta , Methylation , Promoter Regions, Genetic , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Stress, Physiological
11.
Nucleic Acids Res ; 39(4): 1408-18, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20952408

ABSTRACT

Rhesus macaque is a widely used primate model organism. Its genome annotations are however still largely comparative computational predictions derived mainly from human genes, which precludes studies on the macaque-specific genes, gene isoforms or their regulations. Here we took advantage of histone H3 lysine 4 trimethylation (H3K4me3)'s ability to mark transcription start sites (TSSs) and the recently developed ChIP-Seq and RNA-Seq technology to survey the transcript structures. We generated 14,013,757 sequence tags by H3K4me3 ChIP-Seq and obtained 17,322,358 paired end reads for mRNA, and 10,698,419 short reads for sRNA from the macaque brain. By integrating these data with genomic sequence features and extending and improving a state-of-the-art TSS prediction algorithm, we ab initio predicted and verified 17,933 of previously electronically annotated TSSs at 500-bp resolution. We also predicted approximately 10,000 novel TSSs. These provide an important rich resource for close examination of the species-specific transcript structures and transcription regulations in the Rhesus macaque genome. Our approach exemplifies a relatively inexpensive way to generate a reasonably reliable TSS map for a large genome. It may serve as a guiding example for similar genome annotation efforts targeted at other model organisms.


Subject(s)
Genomics/methods , Histones/metabolism , Macaca mulatta/genetics , Transcription Initiation Site , Algorithms , Animals , Chromatin Immunoprecipitation , Genome , High-Throughput Nucleotide Sequencing , Histones/chemistry , Lysine/metabolism , Macaca mulatta/metabolism , Molecular Sequence Annotation , RNA, Messenger/chemistry , RNA, Small Untranslated/chemistry , ROC Curve , Sequence Analysis, RNA
12.
J Genet Genomics ; 35(10): 577-84, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18937914

ABSTRACT

Organism development is a systems level process. It has benefited greatly from the recent technological advances in the field of systems biology. DNA microarray, phenome, interactome and transcriptome mapping, the new generation of deep sequencing technologies, and faster and better computational and modeling approaches have opened new frontiers for both systems biologists and developmental biologists to reexamine the old developmental biology questions, such as pattern formation, and to tackle new problems, such as stem cell reprogramming. As showcased in the International Developmental Systems Biology Symposium organized by Chinese Academy of Sciences, developmental systems biology is flourishing in many perspectives, from the evolution of developmental systems, to the underlying genetic and molecular pathways and networks, to the genomic, epigenomic and noncoding levels, to the computational analysis and modeling. We believe that the field will continue to reap rewards into the future with these new approaches.


Subject(s)
Developmental Biology/methods , Systems Biology/methods , Animals , Computational Biology , Epigenesis, Genetic , Evolution, Molecular , Genomics , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...