Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 89: 283-308, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569523

ABSTRACT

We have known for decades that long noncoding RNAs (lncRNAs) can play essential functions across most forms of life. The maintenance of chromosome length requires an lncRNA (e.g., hTERC) and two lncRNAs in the ribosome that are required for protein synthesis. Thus, lncRNAs can represent powerful RNA machines. More recently, it has become clear that mammalian genomes encode thousands more lncRNAs. Thus, we raise the question: Which, if any, of these lncRNAs could also represent RNA-based machines? Here we synthesize studies that are beginning to address this question by investigating fundamental properties of lncRNA genes, revealing new insights into the RNA structure-function relationship, determining cis- and trans-acting lncRNAs in vivo, and generating new developments in high-throughput screening used to identify functional lncRNAs. Overall, these findings provide a context toward understanding the molecular grammar underlying lncRNA biology.


Subject(s)
Genome , Protein Biosynthesis , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA/genetics , Telomerase/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Humans , Nucleic Acid Conformation , Promoter Regions, Genetic , RNA/metabolism , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Structure-Activity Relationship , Telomerase/metabolism , Telomere Homeostasis , Transcription, Genetic
2.
Nat Rev Mol Cell Biol ; 24(6): 430-447, 2023 06.
Article in English | MEDLINE | ID: mdl-36596869

ABSTRACT

Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I (Pol I), Pol II and Pol III transcribed RNAs, and RNAs from processed introns. The various functions of lncRNAs and their many isoforms and interleaved relationships with other genes make lncRNA classification and annotation difficult. Most lncRNAs evolve more rapidly than protein-coding sequences, are cell type specific and regulate many aspects of cell differentiation and development and other physiological processes. Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates and domains, indicating an intimate link between lncRNA expression and the spatial control of gene expression during development. lncRNAs also have important roles in the cytoplasm and beyond, including in the regulation of translation, metabolism and signalling. lncRNAs often have a modular structure and are rich in repeats, which are increasingly being shown to be relevant to their function. In this Consensus Statement, we address the definition and nomenclature of lncRNAs and their conservation, expression, phenotypic visibility, structure and functions. We also discuss research challenges and provide recommendations to advance the understanding of the roles of lncRNAs in development, cell biology and disease.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , Cell Nucleus/genetics , Chromatin/genetics , Regulatory Sequences, Nucleic Acid , RNA Polymerase II/genetics
3.
Cell ; 183(5): 1383-1401.e19, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33159858

ABSTRACT

Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions/genetics , Single-Cell Analysis , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Bystander Effect , Cell Differentiation , Cell Proliferation , Cytokines/metabolism , Ebolavirus/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Viral , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Histocompatibility Antigens Class II/metabolism , Interferons/genetics , Interferons/metabolism , Macaca mulatta , Macrophages/metabolism , Monocytes/metabolism , Myelopoiesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transcriptome/genetics
4.
Cell ; 173(3): 649-664.e20, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677511

ABSTRACT

Resistance to chemotherapy plays a significant role in cancer mortality. To identify genetic units affecting sensitivity to cytarabine, the mainstay of treatment for acute myeloid leukemia (AML), we developed a comprehensive and integrated genome-wide platform based on a dual protein-coding and non-coding integrated CRISPRa screening (DICaS). Putative resistance genes were initially identified using pharmacogenetic data from 760 human pan-cancer cell lines. Subsequently, genome scale functional characterization of both coding and long non-coding RNA (lncRNA) genes by CRISPR activation was performed. For lncRNA functional assessment, we developed a CRISPR activation of lncRNA (CaLR) strategy, targeting 14,701 lncRNA genes. Computational and functional analysis identified novel cell-cycle, survival/apoptosis, and cancer signaling genes. Furthermore, transcriptional activation of the GAS6-AS2 lncRNA, identified in our analysis, leads to hyperactivation of the GAS6/TAM pathway, a resistance mechanism in multiple cancers including AML. Thus, DICaS represents a novel and powerful approach to identify integrated coding and non-coding pathways of therapeutic relevance.


Subject(s)
CRISPR-Cas Systems , Drug Resistance, Neoplasm , Genome, Human , RNA, Long Noncoding/genetics , Animals , Cytarabine/pharmacology , Female , Gene Expression Profiling , Gene Regulatory Networks , HEK293 Cells , HL-60 Cells , Humans , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Mice , Pharmacogenetics , Proteins/genetics , RNA/analysis , RNA, Messenger/genetics , Signal Transduction
5.
Cell ; 162(2): 412-424, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186193

ABSTRACT

Induced pluripotency is a promising avenue for disease modeling and therapy, but the molecular principles underlying this process, particularly in human cells, remain poorly understood due to donor-to-donor variability and intercellular heterogeneity. Here, we constructed and characterized a clonal, inducible human reprogramming system that provides a reliable source of cells at any stage of the process. This system enabled integrative transcriptional and epigenomic analysis across the human reprogramming timeline at high resolution. We observed distinct waves of gene network activation, including the ordered re-activation of broad developmental regulators followed by early embryonic patterning genes and culminating in the emergence of a signature reminiscent of pre-implantation stages. Moreover, complementary functional analyses allowed us to identify and validate novel regulators of the reprogramming process. Altogether, this study sheds light on the molecular underpinnings of induced pluripotency in human cells and provides a robust cell platform for further studies. PAPERCLIP.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Chromatin/metabolism , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Profiling , Histone Demethylases/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism
6.
Nat Rev Genet ; 23(4): 229-243, 2022 04.
Article in English | MEDLINE | ID: mdl-34837040

ABSTRACT

Genome-wide sequencing has led to the discovery of thousands of long non-coding RNA (lncRNA) loci in the human genome, but evidence of functional significance has remained controversial for many lncRNAs. Genetically engineered model organisms are considered the gold standard for linking genotype to phenotype. Recent advances in CRISPR-Cas genome editing have led to a rapid increase in the use of mouse models to more readily survey lncRNAs for functional significance. Here, we review strategies to investigate the physiological relevance of lncRNA loci by highlighting studies that have used genetic mouse models to reveal key in vivo roles for lncRNAs, from fertility to brain development. We illustrate how an investigative approach, starting with whole-gene deletion followed by transcription termination and/or transgene rescue strategies, can provide definitive evidence for the in vivo function of mammalian lncRNAs.


Subject(s)
RNA, Long Noncoding , Animals , Gene Editing , Genome, Human , Genotype , Humans , Mammals/genetics , Mice , Phenotype , RNA, Long Noncoding/genetics
7.
Cell ; 152(4): 703-13, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23415221

ABSTRACT

Although several hundred regions of the human genome harbor signals of positive natural selection, few of the relevant adaptive traits and variants have been elucidated. Using full-genome sequence variation from the 1000 Genomes (1000G) Project and the composite of multiple signals (CMS) test, we investigated 412 candidate signals and leveraged functional annotation, protein structure modeling, epigenetics, and association studies to identify and extensively annotate candidate causal variants. The resulting catalog provides a tractable list for experimental follow-up; it includes 35 high-scoring nonsynonymous variants, 59 variants associated with expression levels of a nearby coding gene or lincRNA, and numerous variants associated with susceptibility to infectious disease and other phenotypes. We experimentally characterized one candidate nonsynonymous variant in Toll-like receptor 5 (TLR5) and show that it leads to altered NF-κB signaling in response to bacterial flagellin. PAPERFLICK:


Subject(s)
Genetic Techniques , Genome, Human , Genome-Wide Association Study , Mutation , Animals , Bacteria/metabolism , Flagellin/metabolism , HapMap Project , Humans , NF-kappa B/metabolism , Quantitative Trait Loci , Regulatory Elements, Transcriptional , Signal Transduction , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism
8.
Annu Rev Biochem ; 81: 145-66, 2012.
Article in English | MEDLINE | ID: mdl-22663078

ABSTRACT

The central dogma of gene expression is that DNA is transcribed into messenger RNAs, which in turn serve as the template for protein synthesis. The discovery of extensive transcription of large RNA transcripts that do not code for proteins, termed long noncoding RNAs (lncRNAs), provides an important new perspective on the centrality of RNA in gene regulation. Here, we discuss genome-scale strategies to discover and characterize lncRNAs. An emerging theme from multiple model systems is that lncRNAs form extensive networks of ribonucleoprotein (RNP) complexes with numerous chromatin regulators and then target these enzymatic activities to appropriate locations in the genome. Consistent with this notion, lncRNAs can function as modular scaffolds to specify higher-order organization in RNP complexes and in chromatin states. The importance of these modes of regulation is underscored by the newly recognized roles of long RNAs for proper gene control across all kingdoms of life.


Subject(s)
Gene Expression Regulation , RNA, Untranslated/metabolism , Animals , Disease/genetics , Genome, Human , Genome-Wide Association Study , Humans , Ribonucleoproteins/metabolism
9.
Immunity ; 47(3): 435-449.e8, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28930659

ABSTRACT

Commitment to the innate lymphoid cell (ILC) lineage is determined by Id2, a transcriptional regulator that antagonizes T and B cell-specific gene expression programs. Yet how Id2 expression is regulated in each ILC subset remains poorly understood. We identified a cis-regulatory element demarcated by a long non-coding RNA (lncRNA) that controls the function and lineage identity of group 1 ILCs, while being dispensable for early ILC development and homeostasis of ILC2s and ILC3s. The locus encoding this lncRNA, which we termed Rroid, directly interacted with the promoter of its neighboring gene, Id2, in group 1 ILCs. Moreover, the Rroid locus, but not the lncRNA itself, controlled the identity and function of ILC1s by promoting chromatin accessibility and deposition of STAT5 at the promoter of Id2 in response to interleukin (IL)-15. Thus, non-coding elements responsive to extracellular cues unique to each ILC subset represent a key regulatory layer for controlling the identity and function of ILCs.


Subject(s)
Gene Expression Regulation , Immunity, Innate/genetics , Lymphocytes/metabolism , RNA, Long Noncoding/genetics , Regulatory Sequences, Nucleic Acid , Animals , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Chromatin Assembly and Disassembly , Female , Gene Expression Profiling , Genetic Loci , Homeostasis , Inhibitor of Differentiation Protein 2/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/immunology , Male , Mice , Promoter Regions, Genetic , STAT5 Transcription Factor/metabolism , Transcription, Genetic
10.
Mol Cell ; 69(6): 1039-1045.e3, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29526697

ABSTRACT

Imaging (fluorescence in situ hybridization [FISH]) and genome-wide chromosome conformation capture (Hi-C) are two major approaches to the study of higher-order genome organization in the nucleus. Intra-chromosomal and inter-chromosomal interactions (referred to as non-homologous chromosomal contacts [NHCCs]) have been observed by several FISH-based studies, but locus-specific NHCCs have not been detected by Hi-C. Due to crosslinking, neither of these approaches assesses spatiotemporal properties. Toward resolving the discrepancies between imaging and Hi-C, we sought to understand the spatiotemporal properties of NHCCs in living cells by CRISPR/Cas9 live-cell imaging (CLING). In mammalian cells, we find that NHCCs are stable and occur as frequently as intra-chromosomal interactions, but NHCCs occur at farther spatial distance that could explain their lack of detection in Hi-C. By revealing the spatiotemporal properties in living cells, our study provides fundamental insights into the biology of NHCCs.


Subject(s)
Chromosomes, Human/genetics , Microscopy, Confocal/methods , Time-Lapse Imaging/methods , Animals , CRISPR-Cas Systems , Cell Line , Chromosomes, Human/metabolism , Female , Gene Editing/methods , Humans , Image Processing, Computer-Assisted/methods , Kinetics , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Nucleic Acid Conformation , Protein Conformation , Retinal Pigment Epithelium/metabolism
11.
Nucleic Acids Res ; 52(10): e48, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38726866

ABSTRACT

Many of the biological functions performed by RNA are mediated by RNA-binding proteins (RBPs), and understanding the molecular basis of these interactions is fundamental to biology. Here, we present massively parallel RNA assay combined with immunoprecipitation (MPRNA-IP) for in vivo high-throughput dissection of RNA-protein interactions and describe statistical models for identifying RNA domains and parsing the structural contributions of RNA. By using custom pools of tens of thousands of RNA sequences containing systematically designed truncations and mutations, MPRNA-IP is able to identify RNA domains, sequences, and secondary structures necessary and sufficient for protein binding in a single experiment. We show that this approach is successful for multiple RNAs of interest, including the long noncoding RNA NORAD, bacteriophage MS2 RNA, and human telomerase RNA, and we use it to interrogate the hitherto unknown sequence or structural RNA-binding preferences of the DNA-looping factor CTCF. By integrating systematic mutation analysis with crosslinking immunoprecipitation, MPRNA-IP provides a novel high-throughput way to elucidate RNA-based mechanisms behind RNA-protein interactions in vivo.


Subject(s)
RNA-Binding Proteins , RNA , Humans , Binding Sites , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Immunoprecipitation , Levivirus/genetics , Levivirus/metabolism , Mutation , Nucleic Acid Conformation , Protein Binding , RNA/metabolism , RNA/chemistry , RNA/genetics , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/chemistry , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Telomerase/metabolism , Telomerase/genetics , Models, Statistical
12.
RNA ; 29(3): 346-360, 2023 03.
Article in English | MEDLINE | ID: mdl-36574982

ABSTRACT

Aberrant DNA methylation is one of the earliest hallmarks of cancer. DNMT1 is responsible for methylating newly replicated DNA, but the precise regulation of DNMT1 to ensure faithful DNA methylation remains poorly understood. A link between RNA and chromatin-associated proteins has recently emerged, and several studies have shown that DNMT1 can be regulated by a variety of RNAs. In this study, we have confirmed that human DNMT1 indeed interacts with multiple RNAs, including its own nuclear mRNA. Unexpectedly, we found that DNMT1 exhibits a strong and specific affinity for GU-rich RNAs that form a pUG-fold, a noncanonical G-quadruplex. We find that pUG-fold-capable RNAs inhibit DNMT1 activity by inhibiting binding of hemimethylated DNA, and we additionally provide evidence for multiple RNA binding modes with DNMT1. Together, our data indicate that a human chromatin-associated protein binds to and is regulated by pUG-fold RNA.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , Nucleic Acid Conformation , RNA , Humans , Chromatin/metabolism , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , RNA/genetics , RNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism
13.
Cell ; 142(3): 409-19, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20673990

ABSTRACT

Recently, more than 1000 large intergenic noncoding RNAs (lincRNAs) have been reported. These RNAs are evolutionarily conserved in mammalian genomes and thus presumably function in diverse biological processes. Here, we report the identification of lincRNAs that are regulated by p53. One of these lincRNAs (lincRNA-p21) serves as a repressor in p53-dependent transcriptional responses. Inhibition of lincRNA-p21 affects the expression of hundreds of gene targets enriched for genes normally repressed by p53. The observed transcriptional repression by lincRNA-p21 is mediated through the physical association with hnRNP-K. This interaction is required for proper genomic localization of hnRNP-K at repressed genes and regulation of p53 mediates apoptosis. We propose a model whereby transcription factors activate lincRNAs that serve as key repressors by physically associating with repressive complexes and modulate their localization to sets of previously active genes.


Subject(s)
Down-Regulation , RNA, Untranslated/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Mice , Molecular Sequence Data , Transcription, Genetic
14.
RNA ; 28(6): 842-853, 2022 06.
Article in English | MEDLINE | ID: mdl-35304421

ABSTRACT

Long noncoding RNAs (lncRNAs) are rapidly evolving and thus typically poorly conserved in their sequences. How these sequence differences affect the characteristics and potential functions of lncRNAs with shared synteny remains unclear. Here we show that the syntenically conserved lncRNA Firre displays distinct expression and localization patterns in human and mouse. Single molecule RNA FISH reveals that in a range of cell lines, mouse Firre (mFirre) is predominantly nuclear, while human FIRRE (hFIRRE) is distributed between the cytoplasm and nucleus. This localization pattern is maintained in human/mouse hybrid cells expressing both human and mouse Firre, implying that the localization of the lncRNA is species autonomous. We find that the majority of hFIRRE transcripts in the cytoplasm are comprised of isoforms that are enriched in RRD repeats. We furthermore determine that in various tissues, mFirre is more highly expressed than its human counterpart. Our data illustrate that the rapid evolution of syntenic lncRNAs can lead to variations in lncRNA localization and abundance, which in turn may result in disparate lncRNA functions even in closely related species.


Subject(s)
RNA, Long Noncoding , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
15.
Mol Cell ; 62(5): 657-64, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259198

ABSTRACT

There is growing evidence that transcription and nuclear organization are tightly linked. Yet, whether transcription of thousands of long noncoding RNAs (lncRNAs) could play a role in this packaging process remains elusive. Although some lncRNAs have been found to have clear roles in nuclear architecture (e.g., FIRRE, NEAT1, XIST, and others), the vast majority remain poorly understood. In this Perspective, we highlight how the act of transcription can affect nuclear architecture. We synthesize several recent findings into a proposed model where the transcription of lncRNAs can serve as guide-posts for shaping genome organization. This model is similar to the game "cat's cradle," where the shape of a string is successively changed by opening up new sites for finger placement. Analogously, transcription of lncRNAs could serve as "grip holds" for nuclear proteins to pull the genome into new positions. This model could explain general lncRNA properties such as low abundance and tissue specificity. Overall, we propose a general framework for how the act of lncRNA transcription could play a role in organizing the 3D genome.


Subject(s)
Cell Nucleus/metabolism , Genome, Human , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , Cell Nucleus/ultrastructure , Chromatin Assembly and Disassembly , Gene Expression Regulation , Genetic Loci , Humans , Models, Genetic , Nuclear Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , RNA, Messenger/biosynthesis
16.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33446502

ABSTRACT

Haematopoiesis relies on tightly controlled gene expression patterns as development proceeds through a series of progenitors. While the regulation of hematopoietic development has been well studied, the role of noncoding elements in this critical process is a developing field. In particular, the discovery of new regulators of lymphopoiesis could have important implications for our understanding of the adaptive immune system and disease. Here we elucidate how a noncoding element is capable of regulating a broadly expressed transcription factor, Ikaros, in a lymphoid lineage-specific manner, such that it imbues Ikaros with the ability to specify the lymphoid lineage over alternate fates. Deletion of the Daedalus locus, which is proximal to Ikaros, led to a severe reduction in early lymphoid progenitors, exerting control over the earliest fate decisions during lymphoid lineage commitment. Daedalus locus deletion led to alterations in Ikaros isoform expression and a significant reduction in Ikaros protein. The Daedalus locus may function through direct DNA interaction as Hi-C analysis demonstrated an interaction between the two loci. Finally, we identify an Ikaros-regulated erythroid-lymphoid checkpoint that is governed by Daedalus in a lymphoid-lineage-specific manner. Daedalus appears to act as a gatekeeper of Ikaros's broad lineage-specifying functions, selectively stabilizing Ikaros activity in the lymphoid lineage and permitting diversion to the erythroid fate in its absence. These findings represent a key illustration of how a transcription factor with broad lineage expression must work in concert with noncoding elements to orchestrate hematopoietic lineage commitment.


Subject(s)
Hematopoiesis/genetics , Ikaros Transcription Factor/genetics , Lymphopoiesis/genetics , RNA, Untranslated/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental/genetics , Mice
17.
J Biol Chem ; 296: 100694, 2021.
Article in English | MEDLINE | ID: mdl-33895132

ABSTRACT

Myosin heavy chain 7b (MYH7b) is an ancient member of the myosin heavy chain motor protein family that is expressed in striated muscles. In mammalian cardiac muscle, MYH7b RNA is expressed along with two other myosin heavy chains, ß-myosin heavy chain (ß-MyHC) and α-myosin heavy chain (α-MyHC). However, unlike ß-MyHC and α-MyHC, which are maintained in a careful balance at the protein level, the MYH7b locus does not produce a full-length protein in the heart due to a posttranscriptional exon-skipping mechanism that occurs in a tissue-specific manner. Whether this locus has a role in the heart beyond producing its intronic microRNA, miR-499, was unclear. Using cardiomyocytes derived from human induced pluripotent stem cells as a model system, we found that the noncoding exon-skipped RNA (lncMYH7b) affects the transcriptional landscape of human cardiomyocytes, independent of miR-499. Specifically, lncMYH7b regulates the ratio of ß-MyHC to α-MyHC, which is crucial for cardiac contractility. We also found that lncMYH7b regulates beat rate and sarcomere formation in cardiomyocytes. This regulation is likely achieved through control of a member of the TEA domain transcription factor family (TEAD3, which is known to regulate ß-MyHC). Therefore, we conclude that this ancient gene has been repurposed by alternative splicing to produce a regulatory long-noncoding RNA in the human heart that affects cardiac myosin composition.


Subject(s)
Cardiac Myosins/metabolism , Myocardium/metabolism , Myosin Heavy Chains/metabolism , RNA, Long Noncoding/genetics , Cardiac Myosins/chemistry , Humans , Induced Pluripotent Stem Cells , MicroRNAs/genetics , Molecular Dynamics Simulation , Myocardium/cytology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/chemistry , Protein Conformation
18.
J Biol Chem ; 296: 100495, 2021.
Article in English | MEDLINE | ID: mdl-33667549

ABSTRACT

Human embryonic stem cells are a type of pluripotent stem cells (hPSCs) that are used to investigate their differentiation into diverse mature cell types for molecular studies. The mechanisms underlying insulin receptor (IR)-mediated signaling in the maintenance of human pluripotent stem cell (hPSC) identity and cell fate specification are not fully understood. Here, we used two independent shRNAs to stably knock down IRs in two hPSC lines that represent pluripotent stem cells and explored the consequences on expression of key proteins in pathways linked to proliferation and differentiation. We consistently observed lowered pAKT in contrast to increased pERK1/2 and a concordant elevation in pluripotency gene expression. ERK2 chromatin immunoprecipitation, luciferase assays, and ERK1/2 inhibitors established direct causality between ERK1/2 and OCT4 expression. Of importance, RNA sequencing analyses indicated a dysregulation of genes involved in cell differentiation and organismal development. Mass spectrometry-based proteomic analyses further confirmed a global downregulation of extracellular matrix proteins. Subsequent differentiation toward the neural lineage reflected alterations in SOX1+PAX6+ neuroectoderm and FOXG1+ cortical neuron marker expression and protein localization. Collectively, our data underscore the role of IR-mediated signaling in maintaining pluripotency, the extracellular matrix necessary for the stem cell niche, and regulating cell fate specification including the neural lineage.


Subject(s)
Human Embryonic Stem Cells/cytology , Neurons/cytology , Pluripotent Stem Cells/cytology , Receptor, Insulin/metabolism , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Human Embryonic Stem Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Phosphorylation , Pluripotent Stem Cells/metabolism , Proteomics/methods , Signal Transduction
19.
EMBO J ; 37(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29335281

ABSTRACT

In the post-genomic era, thousands of putative noncoding regulatory regions have been identified, such as enhancers, promoters, long noncoding RNAs (lncRNAs), and a cadre of small peptides. These ever-growing catalogs require high-throughput assays to test their functionality at scale. Massively parallel reporter assays have greatly enhanced the understanding of noncoding DNA elements en masse Here, we present a massively parallel RNA assay (MPRNA) that can assay 10,000 or more RNA segments for RNA-based functionality. We applied MPRNA to identify RNA-based nuclear localization domains harbored in lncRNAs. We examined a pool of 11,969 oligos densely tiling 38 human lncRNAs that were fused to a cytosolic transcript. After cell fractionation and barcode sequencing, we identified 109 unique RNA regions that significantly enriched this cytosolic transcript in the nucleus including a cytosine-rich motif. These nuclear enrichment sequences are highly conserved and over-represented in global nuclear fractionation sequencing. Importantly, many of these regions were independently validated by single-molecule RNA fluorescence in situ hybridization. Overall, we demonstrate the utility of MPRNA for future investigation of RNA-based functionalities.


Subject(s)
RNA, Long Noncoding/genetics , Cell Nucleus/genetics , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Sequence Analysis, RNA
20.
EMBO J ; 37(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29921581

ABSTRACT

Chromosomes occupy distinct interphase territories in the three-dimensional nucleus. However, how these chromosome territories are arranged relative to one another is poorly understood. Here, we investigated the inter-chromosomal interactions between chromosomes 2q, 12, and 17 in human mesenchymal stem cells (MSCs) and MSC-derived cell types by DNA-FISH We compared our findings in normal karyotypes with a three-generation family harboring a 2q37-deletion syndrome, featuring a heterozygous partial deletion of histone deacetylase 4 (HDAC4) on chr2q37. In normal karyotypes, we detected stable, recurring arrangements and interactions between the three chromosomal territories with a tissue-specific interaction bias at certain loci. These inter-chromosomal interactions were confirmed by Hi-C. Interestingly, the disease-related HDAC4 deletion resulted in displaced inter-chromosomal arrangements and altered interactions between the deletion-affected chromosome 2 and chromosome 12 and/or 17 in 2q37-deletion syndrome patients. Our findings provide evidence for a direct link between a structural chromosomal aberration and altered interphase architecture that results in a nuclear configuration, supporting a possible molecular pathogenesis.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 2/genetics , Gene Deletion , Histone Deacetylases/genetics , Repressor Proteins/genetics , Translocation, Genetic/genetics , Cell Nucleus/genetics , Chromosome Deletion , Humans , In Situ Hybridization, Fluorescence , Interphase/genetics , Mesenchymal Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL