Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179666

ABSTRACT

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Subject(s)
Chromatin , Placenta , Animals , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Evolution, Molecular , Female , Genome , Mammals/metabolism , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nature ; 592(7852): 93-98, 2021 04.
Article in English | MEDLINE | ID: mdl-33568816

ABSTRACT

Long non-coding RNAs (lncRNAs) can be important components in gene-regulatory networks1, but the exact nature and extent of their involvement in human Mendelian disease is largely unknown. Here we show that genetic ablation of a lncRNA locus on human chromosome 2 causes a severe congenital limb malformation. We identified homozygous 27-63-kilobase deletions located 300 kilobases upstream of the engrailed-1 gene (EN1) in patients with a complex limb malformation featuring mesomelic shortening, syndactyly and ventral nails (dorsal dimelia). Re-engineering of the human deletions in mice resulted in a complete loss of En1 expression in the limb and a double dorsal-limb phenotype that recapitulates the human disease phenotype. Genome-wide transcriptome analysis in the developing mouse limb revealed a four-exon-long non-coding transcript within the deleted region, which we named Maenli. Functional dissection of the Maenli locus showed that its transcriptional activity is required for limb-specific En1 activation in cis, thereby fine-tuning the gene-regulatory networks controlling dorso-ventral polarity in the developing limb bud. Its loss results in the En1-related dorsal ventral limb phenotype, a subset of the full En1-associated phenotype. Our findings demonstrate that mutations involving lncRNA loci can result in human Mendelian disease.


Subject(s)
Extremities , Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , RNA, Long Noncoding/genetics , Sequence Deletion/genetics , Transcription, Genetic , Transcriptional Activation/genetics , Animals , Cell Line , Chromatin/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic
3.
Nat Commun ; 11(1): 5823, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33199677

ABSTRACT

MYCN amplification drives one in six cases of neuroblastoma. The supernumerary gene copies are commonly found on highly rearranged, extrachromosomal circular DNA (ecDNA). The exact amplicon structure has not been described thus far and the functional relevance of its rearrangements is unknown. Here, we analyze the MYCN amplicon structure using short-read and Nanopore sequencing and its chromatin landscape using ChIP-seq, ATAC-seq and Hi-C. This reveals two distinct classes of amplicons which explain the regulatory requirements for MYCN overexpression. The first class always co-amplifies a proximal enhancer driven by the noradrenergic core regulatory circuit (CRC). The second class of MYCN amplicons is characterized by high structural complexity, lacks key local enhancers, and instead contains distal chromosomal fragments harboring CRC-driven enhancers. Thus, ectopic enhancer hijacking can compensate for the loss of local gene regulatory elements and explains a large component of the structural diversity observed in MYCN amplification.


Subject(s)
Chromosomes, Human/genetics , Enhancer Elements, Genetic/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Acetylation , Base Sequence , Cell Line, Tumor , DNA Methylation/genetics , DNA, Circular/genetics , Epigenesis, Genetic , Histones/metabolism , Humans , Kaplan-Meier Estimate , Lysine/metabolism , Nanopore Sequencing
4.
Sci Adv ; 6(35): eabb4591, 2020 08.
Article in English | MEDLINE | ID: mdl-32923640

ABSTRACT

Human genome-wide association studies have linked single-nucleotide polymorphisms (SNPs) in NEMP1 (nuclear envelope membrane protein 1) with early menopause; however, it is unclear whether NEMP1 has any role in fertility. We show that whole-animal loss of NEMP1 homologs in Drosophila, Caenorhabditis elegans, zebrafish, and mice leads to sterility or early loss of fertility. Loss of Nemp leads to nuclear shaping defects, most prominently in the germ line. Biochemical, biophysical, and genetic studies reveal that NEMP proteins support the mechanical stiffness of the germline nuclear envelope via formation of a NEMP-EMERIN complex. These data indicate that the germline nuclear envelope has specialized mechanical properties and that NEMP proteins play essential and conserved roles in fertility.

6.
Mol Cell ; 74(6): 1110-1122, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31226276

ABSTRACT

During embryogenesis, precise gene transcription in space and time requires that distal enhancers and promoters communicate by physical proximity within gene regulatory landscapes. To achieve this, regulatory landscapes fold in nuclear space, creating complex 3D structures that influence enhancer-promoter communication and gene expression and that, when disrupted, can cause disease. Here, we provide an overview of how enhancers and promoters construct regulatory landscapes and how multiple scales of 3D chromatin structure sculpt their communication. We focus on emerging views of what enhancer-promoter contacts and chromatin domains physically represent and how two antagonistic fundamental forces-loop extrusion and homotypic attraction-likely form them. We also examine how these same forces spatially separate regulatory landscapes by functional state, thereby creating higher-order compartments that reconfigure during development to enable proper enhancer-promoter communication.


Subject(s)
Chromatin/ultrastructure , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Genome , Promoter Regions, Genetic , Transcription, Genetic , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromatin/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Humans , Molecular Conformation
7.
Genome Res ; 27(7): 1126-1138, 2017 07.
Article in English | MEDLINE | ID: mdl-28424353

ABSTRACT

The 3D organization of the genome changes concomitantly with expression changes during hematopoiesis and immune activation. Studies have focused either on lamina-associated domains (LADs) or on topologically associated domains (TADs), defined by preferential local chromatin interactions, and chromosome compartments, defined as higher-order interactions between TADs sharing functionally similar states. However, few studies have investigated how these affect one another. To address this, we mapped LADs using Lamin B1-DamID during Jurkat T-cell activation, finding significant genome reorganization at the nuclear periphery dominated by release of loci frequently important for T-cell function. To assess how these changes at the nuclear periphery influence wider genome organization, our DamID data sets were contrasted with TADs and compartments. Features of specific repositioning events were then tested by fluorescence in situ hybridization during T-cell activation. First, considerable overlap between TADs and LADs was observed with the TAD repositioning as a unit. Second, A1 and A2 subcompartments are segregated in 3D space through differences in proximity to LADs along chromosomes. Third, genes and a putative enhancer in LADs that were released from the periphery during T-cell activation became preferentially associated with A2 subcompartments and were constrained to the relative proximity of the lamina. Thus, lamina associations influence internal nuclear organization, and changes in LADs during T-cell activation may provide an important additional mode of gene regulation.


Subject(s)
Chromosomes, Human/metabolism , Enhancer Elements, Genetic , Lamin Type B/metabolism , Lymphocyte Activation , Neoplasm Proteins/metabolism , Nuclear Envelope/metabolism , T-Lymphocytes/metabolism , Chromosomes, Human/genetics , Humans , Jurkat Cells , Lamin Type B/genetics , Neoplasm Proteins/genetics , Nuclear Envelope/genetics , T-Lymphocytes/cytology
8.
Neuromuscul Disord ; 27(4): 338-351, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28214269

ABSTRACT

Reports of aberrant distribution for some nuclear envelope proteins in cells expressing a few Emery-Dreifuss muscular dystrophy mutations raised the possibility that such protein redistribution could underlie pathology and/or be diagnostic. However, this disorder is linked to 8 different genes encoding nuclear envelope proteins, raising the question of whether a particular protein is most relevant. Therefore, myoblast/fibroblast cultures from biopsy and tissue sections from a panel of nine Emery-Dreifuss muscular dystrophy patients (4 male, 5 female) including those carrying emerin and FHL1 (X-linked) and several lamin A (autosomal dominant) mutations were stained for the proteins linked to the disorder. As tissue-specific nuclear envelope proteins have been postulated to mediate the tissue-specific pathologies of different nuclear envelopathies, patient samples were also stained for several muscle-specific nuclear membrane proteins. Although linked proteins nesprin 1 and SUN2 and muscle-specific proteins NET5/Samp1 and Tmem214 yielded aberrant distributions in individual patient cells, none exhibited defects through the larger patient panel. Muscle-specific Tmem38A normally appeared in both the nuclear envelope and sarcoplasmic reticulum, but most patient samples exhibited a moderate redistribution favouring the sarcoplasmic reticulum. The absence of striking uniform defects in nuclear envelope protein distribution indicates that such staining will be unavailing for general diagnostics, though it remains possible that specific mutations exhibiting protein distribution defects might reflect a particular clinical variant. These findings further argue that multiple pathways can lead to the generally similar pathologies of this disorder while at the same time the different cellular phenotypes observed possibly may help explain the considerable clinical variation of EDMD.


Subject(s)
Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Tissue Banks , Adolescent , Adult , Biomarkers/metabolism , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Skeletal , Muscular Dystrophy, Emery-Dreifuss/pathology
9.
Mol Cell ; 62(6): 834-847, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27264872

ABSTRACT

Whether gene repositioning to the nuclear periphery during differentiation adds another layer of regulation to gene expression remains controversial. Here, we resolve this by manipulating gene positions through targeting the nuclear envelope transmembrane proteins (NETs) that direct their normal repositioning during myogenesis. Combining transcriptomics with high-resolution DamID mapping of nuclear envelope-genome contacts, we show that three muscle-specific NETs, NET39, Tmem38A, and WFS1, direct specific myogenic genes to the nuclear periphery to facilitate their repression. Retargeting a NET39 fragment to nucleoli correspondingly repositioned a target gene, indicating a direct tethering mechanism. Being able to manipulate gene position independently of other changes in differentiation revealed that repositioning contributes ⅓ to ⅔ of a gene's normal repression in myogenesis. Together, these NETs affect 37% of all genes changing expression during myogenesis, and their combined knockdown almost completely blocks myotube formation. This unequivocally demonstrates that NET-directed gene repositioning is critical for developmental gene regulation.


Subject(s)
Chromosome Positioning , Gene Expression Regulation, Developmental , Ion Channels/genetics , Membrane Proteins/genetics , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Animals , Cell Differentiation , Cell Line , Down-Regulation , Humans , Ion Channels/metabolism , Kinetics , Membrane Proteins/metabolism , Mice , Nuclear Proteins/metabolism , RNA Interference , Transfection
10.
Front Genet ; 7: 82, 2016.
Article in English | MEDLINE | ID: mdl-27200088

ABSTRACT

It is well established that the nuclear envelope has many distinct direct connections to chromatin that contribute to genome organization. The functional consequences of genome organization on gene regulation are less clear. Even less understood is how interactions of lamins and nuclear envelope transmembrane proteins (NETs) with chromatin can produce anchoring tethers that can withstand the physical forces of and on the genome. Chromosomes are the largest molecules in the cell, making megadalton protein structures like the nuclear pore complexes and ribosomes seem small by comparison. Thus to withstand strong forces from chromosome dynamics an anchoring tether is likely to be much more complex than a single protein-protein or protein-DNA interaction. Here we will briefly review known NE-genome interactions that likely contribute to spatial genome organization, postulate in the context of experimental data how these anchoring tethers contribute to gene regulation, and posit several hypotheses for the physical nature of these tethers that need to be investigated experimentally. Significantly, disruption of these anchoring tethers and the subsequent consequences for gene regulation could explain how mutations in nuclear envelope proteins cause diseases ranging from muscular dystrophy to lipodystrophy to premature aging progeroid syndromes. The two favored hypotheses for nuclear envelope protein involvement in disease are (1) weakening nuclear and cellular mechanical stability, and (2) disrupting genome organization and gene regulation. Considerable experimental support has been obtained for both. The integration of both mechanical and gene expression defects in the disruption of anchoring tethers could provide a unifying hypothesis consistent with both.

11.
Methods Mol Biol ; 1411: 359-86, 2016.
Article in English | MEDLINE | ID: mdl-27147054

ABSTRACT

The nuclear envelope interacts extensively with chromatin, though with differences in degree and specificity in different cell types. However, identifying the specific genome sequences associated with individual nuclear envelope associated proteins, particularly nuclear membrane proteins and lamins, has been particularly difficult due to their inherent insolubility and interconnectivity. DamID is a powerful tool developed to bypass many of the inherent difficulties with identifying nuclear envelope protein-chromatin interactions and, as more tissue culture cell types derived from different tissues are examined by DamID, it is increasingly apparent that there are distinct patterns of genome organization in differentiated cell types. However, in applying DamID to both more diverse and/or differentiated cell types a number of technical caveats to the method have been observed which must be circumvented to ensure high quality data is generated. Here we elaborate a detailed methodology to adapt DamID to novel cell types, in particular differentiated cells in culture. Moreover, we highlight heretofore largely ignored variations in the PCR amplified DNA products generated by the DamID procedure and the consequences they have for downstream analysis steps. Thus, the methods described here should serve as a useful resource to researchers new to DamID as well as readily allow its application to an expanded set of cell types and conditions.


Subject(s)
Cell Differentiation/genetics , Chromosome Mapping , Methyltransferases/genetics , Recombinant Fusion Proteins/genetics , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromosome Mapping/methods , Computational Biology/methods , DNA Methylation , Genetic Vectors/genetics , High-Throughput Nucleotide Sequencing , Humans , Lentivirus/genetics , Methyltransferases/metabolism , Mice , Muscle Development/genetics , Myoblasts/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Nucleic Acid Amplification Techniques , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
12.
Genome Res ; 25(4): 478-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644835

ABSTRACT

While analyzing the DNA methylome of multiple myeloma (MM), a plasma cell neoplasm, by whole-genome bisulfite sequencing and high-density arrays, we observed a highly heterogeneous pattern globally characterized by regional DNA hypermethylation embedded in extensive hypomethylation. In contrast to the widely reported DNA hypermethylation of promoter-associated CpG islands (CGIs) in cancer, hypermethylated sites in MM, as opposed to normal plasma cells, were located outside CpG islands and were unexpectedly associated with intronic enhancer regions defined in normal B cells and plasma cells. Both RNA-seq and in vitro reporter assays indicated that enhancer hypermethylation is globally associated with down-regulation of its host genes. ChIP-seq and DNase-seq further revealed that DNA hypermethylation in these regions is related to enhancer decommissioning. Hypermethylated enhancer regions overlapped with binding sites of B cell-specific transcription factors (TFs) and the degree of enhancer methylation inversely correlated with expression levels of these TFs in MM. Furthermore, hypermethylated regions in MM were methylated in stem cells and gradually became demethylated during normal B-cell differentiation, suggesting that MM cells either reacquire epigenetic features of undifferentiated cells or maintain an epigenetic signature of a putative myeloma stem cell progenitor. Overall, we have identified DNA hypermethylation of developmentally regulated enhancers as a new type of epigenetic modification associated with the pathogenesis of MM.


Subject(s)
DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Multiple Myeloma/genetics , Neoplastic Stem Cells/cytology , Plasma Cells/cytology , Cell Differentiation/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA, Neoplasm/genetics , Down-Regulation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Promoter Regions, Genetic , Transcription Factors/biosynthesis , Transcription Factors/genetics
13.
Adv Exp Med Biol ; 773: 165-85, 2014.
Article in English | MEDLINE | ID: mdl-24563348

ABSTRACT

There are many ways that the nuclear envelope can influence the cell cycle. In addition to roles of lamins in regulating the master cell cycle regulator pRb and nuclear envelope breakdown in mitosis, many other nuclear envelope proteins influence the cell cycle through regulatory or structural functions. Of particular note among these are the nuclear envelope transmembrane proteins (NETs) that appear to influence cell cycle regulation through multiple separate mechanisms. Some NETs and other nuclear envelope proteins accumulate on the mitotic spindle, suggesting functional or structural roles in the cell cycle. In interphase exogenous overexpression of some NETs promotes an increase in G1 populations, while others promote an increase in G2/M populations, sometimes associated with the induction of senescence. Intriguingly, most of the NETs linked to the cell cycle are highly restricted in their tissue expression; thus, their misregulation in cancer could contribute to the many tissue-specific types of cancer.


Subject(s)
Cell Cycle , Membrane Proteins/metabolism , Neoplasms/pathology , Nuclear Envelope/metabolism , Humans , Neoplasms/metabolism
14.
Nucleus ; 2(5): 339-49, 2011.
Article in English | MEDLINE | ID: mdl-21970986

ABSTRACT

In the past 15 years our perception of nuclear envelope function has evolved perhaps nearly as much as the nuclear envelope itself evolved in the last 3 billion years. Historically viewed as little more than a diffusion barrier between the cytoplasm and the nucleoplasm, the nuclear envelope is now known to have roles in the cell cycle, cytoskeletal stability and cell migration, genome architecture, epigenetics, regulation of transcription, splicing, and DNA replication. Here we will review both what is known and what is speculated about the role of the nuclear envelope in genome organization, particularly with respect to the positioning and repositioning of genes and chromosomes within the nucleus during differentiation.


Subject(s)
Chromatin/metabolism , Nuclear Envelope/metabolism , Cell Nucleus/physiology , Chromosomes/physiology , Nuclear Pore/physiology , Nuclear Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...