Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.865
Filter
1.
Nat Immunol ; 24(7): 1188-1199, 2023 07.
Article in English | MEDLINE | ID: mdl-37322178

ABSTRACT

Spalt-like transcription factor 1 (SALL1) is a critical regulator of organogenesis and microglia identity. Here we demonstrate that disruption of a conserved microglia-specific super-enhancer interacting with the Sall1 promoter results in complete and specific loss of Sall1 expression in microglia. By determining the genomic binding sites of SALL1 and leveraging Sall1 enhancer knockout mice, we provide evidence for functional interactions between SALL1 and SMAD4 required for microglia-specific gene expression. SMAD4 binds directly to the Sall1 super-enhancer and is required for Sall1 expression, consistent with an evolutionarily conserved requirement of the TGFß and SMAD homologs Dpp and Mad for cell-specific expression of Spalt in the Drosophila wing. Unexpectedly, SALL1 in turn promotes binding and function of SMAD4 at microglia-specific enhancers while simultaneously suppressing binding of SMAD4 to enhancers of genes that become inappropriately activated in enhancer knockout microglia, thereby enforcing microglia-specific functions of the TGFß-SMAD signaling axis.


Subject(s)
Microglia , Transcription Factors , Animals , Mice , Binding Sites , DNA , Mice, Knockout , Microglia/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
2.
Cell ; 181(6): 1410-1422.e27, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32413320

ABSTRACT

Tracing the lineage history of cells is key to answering diverse and fundamental questions in biology. Coupling of cell ancestry information with other molecular readouts represents an important goal in the field. Here, we describe the CRISPR array repair lineage tracing (CARLIN) mouse line and corresponding analysis tools that can be used to simultaneously interrogate the lineage and transcriptomic information of single cells in vivo. This model exploits CRISPR technology to generate up to 44,000 transcribed barcodes in an inducible fashion at any point during development or adulthood, is compatible with sequential barcoding, and is fully genetically defined. We have used CARLIN to identify intrinsic biases in the activity of fetal liver hematopoietic stem cell (HSC) clones and to uncover a previously unappreciated clonal bottleneck in the response of HSCs to injury. CARLIN also allows the unbiased identification of transcriptional signatures associated with HSC activity without cell sorting.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Transcriptome/genetics , Animals , Cell Line , Female , Flow Cytometry/methods , Hematopoietic Stem Cells/physiology , Male , Mice , Transduction, Genetic/methods
3.
Nat Immunol ; 22(9): 1175-1185, 2021 09.
Article in English | MEDLINE | ID: mdl-34429546

ABSTRACT

Systematic characterizations of adipose regulatory T (Treg) cell subsets and their phenotypes remain uncommon. Using single-cell ATAC-sequencing and paired single-cell RNA and T cell receptor (TCR) sequencing to map mouse adipose Treg cells, we identified CD73hiST2lo and CD73loST2hi subsets with distinct clonal expansion patterns. Analysis of TCR-sharing data implied a state transition between CD73hiST2lo and CD73loST2hi subsets. Mechanistically, we revealed that insulin signaling occurs through a HIF-1α-Med23-PPAR-γ axis to drive the transition of CD73hiST2lo into a CD73loST2hi adipose Treg cell subset. Treg cells deficient in insulin receptor, HIF-1α or Med23 have decreased PPAR-γ expression that in turn promotes accumulation of CD73hiST2lo adipose Treg cells and physiological adenosine production to activate beige fat biogenesis. We therefore unveiled a developmental trajectory of adipose Treg cells and its dependence on insulin signaling. Our findings have implications for understanding the dynamics of adipose Treg cell subsets in aged and obese contexts.


Subject(s)
Adipose Tissue/immunology , Insulin Resistance/immunology , Insulin/metabolism , Receptor, Insulin/metabolism , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adipose Tissue/cytology , Aging/immunology , Animals , Cells, Cultured , High-Throughput Nucleotide Sequencing , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Male , Mediator Complex/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/immunology , PPAR gamma/metabolism , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology
4.
Cell ; 175(6): 1607-1619.e15, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500539

ABSTRACT

In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in vitro from bile-duct epithelial cells, but not hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6 months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah-/- mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Culture Techniques , Cell Proliferation/drug effects , Hepatocytes/metabolism , Spheroids, Cellular/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line, Transformed , Hep G2 Cells , Hepatocytes/transplantation , Human Umbilical Vein Endothelial Cells , Humans , Liver/injuries , Liver/metabolism , Mice, Knockout , Spheroids, Cellular/transplantation , Time Factors
5.
Cell ; 166(4): 881-893, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27518563

ABSTRACT

Classically, hormones elicit specific cellular responses by activating dedicated receptors. Nevertheless, the biosynthesis and turnover of many of these hormone molecules also produce chemically related metabolites. These molecules may also possess hormonal activities; therefore, one or more may contribute to the adaptive plasticity of signaling outcomes in host organisms. Here, we show that a catabolite of the plant hormone abscisic acid (ABA), namely phaseic acid (PA), likely emerged in seed plants as a signaling molecule that fine-tunes plant physiology, environmental adaptation, and development. This trait was facilitated by both the emergence-selection of a PA reductase that modulates PA concentrations and by the functional diversification of the ABA receptor family to perceive and respond to PA. Our results suggest that PA serves as a hormone in seed plants through activation of a subset of ABA receptors. This study demonstrates that the co-evolution of hormone metabolism and signaling networks can expand organismal resilience.


Subject(s)
Abscisic Acid/metabolism , Arabidopsis/metabolism , Plant Growth Regulators/metabolism , Sesquiterpenes/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , NADP/metabolism , Signal Transduction
7.
Nature ; 627(8005): 854-864, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480880

ABSTRACT

The heart, which is the first organ to develop, is highly dependent on its form to function1,2. However, how diverse cardiac cell types spatially coordinate to create the complex morphological structures that are crucial for heart function remains unclear. Here we integrated single-cell RNA-sequencing with high-resolution multiplexed error-robust fluorescence in situ hybridization to resolve the identity of the cardiac cell types that develop the human heart. This approach also provided a spatial mapping of individual cells that enables illumination of their organization into cellular communities that form distinct cardiac structures. We discovered that many of these cardiac cell types further specified into subpopulations exclusive to specific communities, which support their specialization according to the cellular ecosystem and anatomical region. In particular, ventricular cardiomyocyte subpopulations displayed an unexpected complex laminar organization across the ventricular wall and formed, with other cell subpopulations, several cellular communities. Interrogating cell-cell interactions within these communities using in vivo conditional genetic mouse models and in vitro human pluripotent stem cell systems revealed multicellular signalling pathways that orchestrate the spatial organization of cardiac cell subpopulations during ventricular wall morphogenesis. These detailed findings into the cellular social interactions and specialization of cardiac cell types constructing and remodelling the human heart offer new insights into structural heart diseases and the engineering of complex multicellular tissues for human heart repair.


Subject(s)
Body Patterning , Heart , Myocardium , Animals , Humans , Mice , Heart/anatomy & histology , Heart/embryology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/embryology , In Situ Hybridization, Fluorescence , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Gene Expression Analysis
8.
Nature ; 624(7991): 378-389, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092917

ABSTRACT

Recent advances in single-cell technologies have led to the discovery of thousands of brain cell types; however, our understanding of the gene regulatory programs in these cell types is far from complete1-4. Here we report a comprehensive atlas of candidate cis-regulatory DNA elements (cCREs) in the adult mouse brain, generated by analysing chromatin accessibility in 2.3 million individual brain cells from 117 anatomical dissections. The atlas includes approximately 1 million cCREs and their chromatin accessibility across 1,482 distinct brain cell populations, adding over 446,000 cCREs to the most recent such annotation in the mouse genome. The mouse brain cCREs are moderately conserved in the human brain. The mouse-specific cCREs-specifically, those identified from a subset of cortical excitatory neurons-are strongly enriched for transposable elements, suggesting a potential role for transposable elements in the emergence of new regulatory programs and neuronal diversity. Finally, we infer the gene regulatory networks in over 260 subclasses of mouse brain cells and develop deep-learning models to predict the activities of gene regulatory elements in different brain cell types from the DNA sequence alone. Our results provide a resource for the analysis of cell-type-specific gene regulation programs in both mouse and human brains.


Subject(s)
Brain , Chromatin , Single-Cell Analysis , Animals , Humans , Mice , Brain/cytology , Brain/metabolism , Cerebral Cortex/cytology , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Deep Learning , DNA Transposable Elements/genetics , Gene Regulatory Networks/genetics , Neurons/metabolism
9.
Nature ; 612(7940): 519-527, 2022 12.
Article in English | MEDLINE | ID: mdl-36477534

ABSTRACT

In mice and humans, sleep quantity is governed by genetic factors and exhibits age-dependent variation1-3. However, the core molecular pathways and effector mechanisms that regulate sleep duration in mammals remain unclear. Here, we characterize a major signalling pathway for the transcriptional regulation of sleep in mice using adeno-associated virus-mediated somatic genetics analysis4. Chimeric knockout of LKB1 kinase-an activator of AMPK-related protein kinase SIK35-7-in adult mouse brain markedly reduces the amount and delta power-a measure of sleep depth-of non-rapid eye movement sleep (NREMS). Downstream of the LKB1-SIK3 pathway, gain or loss-of-function of the histone deacetylases HDAC4 and HDAC5 in adult brain neurons causes bidirectional changes of NREMS amount and delta power. Moreover, phosphorylation of HDAC4 and HDAC5 is associated with increased sleep need, and HDAC4 specifically regulates NREMS amount in posterior hypothalamus. Genetic and transcriptomic studies reveal that HDAC4 cooperates with CREB in both transcriptional and sleep regulation. These findings introduce the concept of signalling pathways targeting transcription modulators to regulate daily sleep amount and demonstrate the power of somatic genetics in mouse sleep research.


Subject(s)
Signal Transduction , Sleep Duration , Transcription, Genetic , Animals , Mice , Gene Expression Regulation , Phosphorylation , Signal Transduction/physiology , Sleep, Slow-Wave/genetics , Gene Expression Profiling
10.
Nature ; 599(7884): 325-329, 2021 11.
Article in English | MEDLINE | ID: mdl-34552241

ABSTRACT

Glutamate-gated kainate receptors are ubiquitous in the central nervous system of vertebrates, mediate synaptic transmission at the postsynapse and modulate transmitter release at the presynapse1-7. In the brain, the trafficking, gating kinetics and pharmacology of kainate receptors are tightly regulated by neuropilin and tolloid-like (NETO) proteins8-11. Here we report cryo-electron microscopy structures of homotetrameric GluK2 in complex with NETO2 at inhibited and desensitized states, illustrating variable stoichiometry of GluK2-NETO2 complexes, with one or two NETO2 subunits associating with GluK2. We find that NETO2 accesses only two broad faces of kainate receptors, intermolecularly crosslinking the lower lobe of ATDA/C, the upper lobe of LBDB/D and the lower lobe of LBDA/C, illustrating how NETO2 regulates receptor-gating kinetics. The transmembrane helix of NETO2 is positioned proximal to the selectivity filter and competes with the amphiphilic H1 helix after M4 for interaction with an intracellular cap domain formed by the M1-M2 linkers of the receptor, revealing how rectification is regulated by NETO2.


Subject(s)
Membrane Proteins/metabolism , Receptors, Kainic Acid/metabolism , Cryoelectron Microscopy , Electrophysiology , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Models, Molecular , Protein Binding , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/ultrastructure , GluK2 Kainate Receptor
11.
Nature ; 598(7879): 129-136, 2021 10.
Article in English | MEDLINE | ID: mdl-34616068

ABSTRACT

The mammalian cerebrum performs high-level sensory perception, motor control and cognitive functions through highly specialized cortical and subcortical structures1. Recent surveys of mouse and human brains with single-cell transcriptomics2-6 and high-throughput imaging technologies7,8 have uncovered hundreds of neural cell types distributed in different brain regions, but the transcriptional regulatory programs that are responsible for the unique identity and function of each cell type remain unknown. Here we probe the accessible chromatin in more than 800,000 individual nuclei from 45 regions that span the adult mouse isocortex, olfactory bulb, hippocampus and cerebral nuclei, and use the resulting data to map the state of 491,818 candidate cis-regulatory DNA elements in 160 distinct cell types. We find high specificity of spatial distribution for not only excitatory neurons, but also most classes of inhibitory neurons and a subset of glial cell types. We characterize the gene regulatory sequences associated with the regional specificity within these cell types. We further link a considerable fraction of the cis-regulatory elements to putative target genes expressed in diverse cerebral cell types and predict transcriptional regulators that are involved in a broad spectrum of molecular and cellular pathways in different neuronal and glial cell populations. Our results provide a foundation for comprehensive analysis of gene regulatory programs of the mammalian brain and assist in the interpretation of noncoding risk variants associated with various neurological diseases and traits in humans.


Subject(s)
Cerebrum/cytology , Cerebrum/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Atlases as Topic , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Gene Expression Regulation , Genetic Predisposition to Disease/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Nervous System Diseases/genetics , Neuroglia/classification , Neuroglia/metabolism , Neurons/classification , Neurons/metabolism , Sequence Analysis, DNA , Single-Cell Analysis
12.
N Engl J Med ; 388(20): 1843-1852, 2023 May 18.
Article in English | MEDLINE | ID: mdl-37195940

ABSTRACT

BACKGROUND: Previous studies have suggested that a single dose of rifampin has protective effects against leprosy in close contacts of patients with the disease. Rifapentine was shown to have greater bactericidal activity against Mycobacterium leprae than rifampin in murine models of leprosy, but data regarding its effectiveness in preventing leprosy are lacking. METHODS: We conducted a cluster-randomized, controlled trial to investigate whether single-dose rifapentine is effective in preventing leprosy in household contacts of patients with leprosy. The clusters (counties or districts in Southwest China) were assigned to one of three trial groups: single-dose rifapentine, single-dose rifampin, or control (no intervention). The primary outcome was the 4-year cumulative incidence of leprosy among household contacts. RESULTS: A total of 207 clusters comprising 7450 household contacts underwent randomization; 68 clusters (2331 household contacts) were assigned to the rifapentine group, 71 (2760) to the rifampin group, and 68 (2359) to the control group. A total of 24 new cases of leprosy occurred over the 4-year follow-up, for a cumulative incidence of 0.09% (95% confidence interval [CI], 0.02 to 0.34) with rifapentine (2 cases), 0.33% (95% CI, 0.17 to 0.63) with rifampin (9 cases), and 0.55% (95% CI, 0.32 to 0.95) with no intervention (13 cases). In an intention-to-treat analysis, the cumulative incidence in the rifapentine group was 84% lower than that in the control group (cumulative incidence ratio, 0.16; multiplicity-adjusted 95% CI, 0.03 to 0.87; P = 0.02); the cumulative incidence did not differ significantly between the rifampin group and the control group (cumulative incidence ratio, 0.59; multiplicity-adjusted 95% CI, 0.22 to 1.57; P = 0.23). In a per-protocol analysis, the cumulative incidence was 0.05% with rifapentine, 0.19% with rifampin, and 0.63% with no intervention. No severe adverse events were observed. CONCLUSIONS: The incidence of leprosy among household contacts over 4 years was lower with single-dose rifapentine than with no intervention. (Funded by the Ministry of Health of China and the Chinese Academy of Medical Sciences; Chinese Clinical Trial Registry number, ChiCTR-IPR-15007075.).


Subject(s)
Leprostatic Agents , Leprosy , Mycobacterium leprae , Rifampin , Humans , Incidence , Leprosy/epidemiology , Leprosy/prevention & control , Leprosy/transmission , Rifampin/administration & dosage , Rifampin/analogs & derivatives , Leprostatic Agents/administration & dosage , Leprostatic Agents/therapeutic use , Family Characteristics
13.
PLoS Biol ; 21(3): e3002039, 2023 03.
Article in English | MEDLINE | ID: mdl-36930652

ABSTRACT

Coronaviruses (CoVs) comprise a group of important human and animal pathogens. Despite extensive research in the past 3 years, the host innate immune defense mechanisms against CoVs remain incompletely understood, limiting the development of effective antivirals and non-antibody-based therapeutics. Here, we performed an integrated transcriptomic analysis of porcine jejunal epithelial cells infected with porcine epidemic diarrhea virus (PEDV) and identified cytidine/uridine monophosphate kinase 2 (CMPK2) as a potential host restriction factor. CMPK2 exhibited modest antiviral activity against PEDV infection in multiple cell types. CMPK2 transcription was regulated by interferon-dependent and interferon regulatory factor 1 (IRF1)-dependent pathways post-PEDV infection. We demonstrated that 3'-deoxy-3',4'-didehydro-cytidine triphosphate (ddhCTP) catalysis by Viperin, another interferon-stimulated protein, was essential for CMPK2's antiviral activity. Both the classical catalytic domain and the newly identified antiviral key domain of CMPK2 played crucial roles in this process. Together, CMPK2, viperin, and ddhCTP suppressed the replication of several other CoVs of different genera through inhibition of the RNA-dependent RNA polymerase activities. Our results revealed a previously unknown function of CMPK2 as a restriction factor for CoVs, implying that CMPK2 might be an alternative target of interfering with the viral polymerase activity.


Subject(s)
Coronavirus Infections , Coronavirus , Porcine epidemic diarrhea virus , Humans , Animals , Swine , Interferons , Antiviral Agents/pharmacology , Proteins/genetics , Porcine epidemic diarrhea virus/genetics
14.
EMBO Rep ; 25(1): 128-143, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177907

ABSTRACT

Collateral circulation is essential for blood resupply to the ischemic heart, which is dictated by the contractile phenotypic restoration of vascular smooth muscle cells (VSMC). Here we investigate whether S-nitrosylation of AMP-activated protein kinase (AMPK), a key regulator of the VSMC phenotype, impairs collateral circulation. In rats with collateral growth and development, nitroglycerin decreases coronary collateral blood flow (CCBF), inhibits vascular contractile phenotypic restoration, and increases myocardial infarct size, accompanied by reduced AMPK activity in the collateral zone. Nitric oxide (NO) S-nitrosylates human recombinant AMPKγ1 at cysteine 131 and decreases AMP sensitivity of AMPK. In VSMCs, exogenous expression of S-nitrosylation-resistant AMPKγ1 or deficient NO synthase (iNOS) prevents the disruption of VSMC reprogramming. Finally, hyperhomocysteinemia or hyperglycemia increases AMPKγ1 S-nitrosylation, prevents vascular contractile phenotypic restoration, reduces CCBF, and increases the infarct size of the heart in Apoe-/- mice, all of which is rescued in Apoe-/-/iNOSsm-/- mice or Apoe-/- mice with enforced expression of the AMPKγ1-C130A mutant following RI/MI. We conclude that nitrosative stress disrupts coronary collateral circulation during hyperhomocysteinemia or hyperglycemia through AMPK S-nitrosylation.


Subject(s)
Hyperglycemia , Hyperhomocysteinemia , Rats , Mice , Humans , Animals , Collateral Circulation , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular , Hyperhomocysteinemia/metabolism , Apolipoproteins E/metabolism , Hyperglycemia/metabolism
15.
EMBO Rep ; 25(2): 770-795, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182816

ABSTRACT

DExD/H-box helicases are crucial regulators of RNA metabolism and antiviral innate immune responses; however, their role in bacteria-induced inflammation remains unclear. Here, we report that DDX5 interacts with METTL3 and METTL14 to form an m6A writing complex, which adds N6-methyladenosine to transcripts of toll-like receptor (TLR) 2 and TLR4, promoting their decay via YTHDF2-mediated RNA degradation, resulting in reduced expression of TLR2/4. Upon bacterial infection, DDX5 is recruited to Hrd1 at the endoplasmic reticulum in an MyD88-dependent manner and is degraded by the ubiquitin-proteasome pathway. This process disrupts the DDX5 m6A writing complex and halts m6A modification as well as degradation of TLR2/4 mRNAs, thereby promoting the expression of TLR2 and TLR4 and downstream NF-κB activation. The role of DDX5 in regulating inflammation is also validated in vivo, as DDX5- and METTL3-KO mice exhibit enhanced expression of inflammatory cytokines. Our findings show that DDX5 acts as a molecular switch to regulate inflammation during bacterial infection and shed light on mechanisms of quiescent inflammation during homeostasis.


Subject(s)
Adenine , Bacterial Infections , Toll-Like Receptor 2 , Animals , Mice , Adenine/analogs & derivatives , Inflammation/genetics , Methyltransferases/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
16.
Nature ; 577(7791): 492-496, 2020 01.
Article in English | MEDLINE | ID: mdl-31969724

ABSTRACT

Although two-dimensional (2D) atomic layers, such as transition-metal chalcogenides, have been widely synthesized using techniques such as exfoliation1-3 and vapour-phase growth4,5, it is still challenging to obtain phase-controlled 2D structures6-8. Here we demonstrate an effective synthesis strategy via the progressive transformation of non-van der Waals (non-vdW) solids to 2D vdW transition-metal chalcogenide layers with identified 2H (trigonal prismatic)/1T (octahedral) phases. The transformation, achieved by exposing non-vdW solids to chalcogen vapours, can be controlled using the enthalpies and vapour pressures of the reaction products. Heteroatom-substituted (such as yttrium and phosphorus) transition-metal chalcogenides can also be synthesized in this way, thus enabling a generic synthesis approach to engineering phase-selected 2D transition-metal chalcogenide structures with good stability at high temperatures (up to 1,373 kelvin) and achieving high-throughput production of monolayers. We anticipate that these 2D transition-metal chalcogenides will have broad applications for electronics, catalysis and energy storage.

17.
Nature ; 583(7818): 752-759, 2020 07.
Article in English | MEDLINE | ID: mdl-32728242

ABSTRACT

Cytosine DNA methylation is essential for mammalian development but understanding of its spatiotemporal distribution in the developing embryo remains limited1,2. Here, as part of the mouse Encyclopedia of DNA Elements (ENCODE) project, we profiled 168 methylomes from 12 mouse tissues or organs at 9 developmental stages from embryogenesis to adulthood. We identified 1,808,810 genomic regions that showed variations in CG methylation by comparing the methylomes of different tissues or organs from different developmental stages. These DNA elements predominantly lose CG methylation during fetal development, whereas the trend is reversed after birth. During late stages of fetal development, non-CG methylation accumulated within the bodies of key developmental transcription factor genes, coinciding with their transcriptional repression. Integration of genome-wide DNA methylation, histone modification and chromatin accessibility data enabled us to predict 461,141 putative developmental tissue-specific enhancers, the human orthologues of which were enriched for disease-associated genetic variants. These spatiotemporal epigenome maps provide a resource for studies of gene regulation during tissue or organ progression, and a starting point for investigating regulatory elements that are involved in human developmental disorders.


Subject(s)
DNA Methylation , Epigenome , Fetus/embryology , Fetus/metabolism , Animals , Animals, Newborn , Chromatin/genetics , Chromatin/metabolism , Disease/genetics , Down-Regulation , Enhancer Elements, Genetic/genetics , Epigenetic Repression , Female , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Models, Animal , Molecular Sequence Annotation , Polymorphism, Single Nucleotide , Spatio-Temporal Analysis
18.
Nature ; 583(7818): 744-751, 2020 07.
Article in English | MEDLINE | ID: mdl-32728240

ABSTRACT

The Encyclopedia of DNA Elements (ENCODE) project has established a genomic resource for mammalian development, profiling a diverse panel of mouse tissues at 8 developmental stages from 10.5 days after conception until birth, including transcriptomes, methylomes and chromatin states. Here we systematically examined the state and accessibility of chromatin in the developing mouse fetus. In total we performed 1,128 chromatin immunoprecipitation with sequencing (ChIP-seq) assays for histone modifications and 132 assay for transposase-accessible chromatin using sequencing (ATAC-seq) assays for chromatin accessibility across 72 distinct tissue-stages. We used integrative analysis to develop a unified set of chromatin state annotations, infer the identities of dynamic enhancers and key transcriptional regulators, and characterize the relationship between chromatin state and accessibility during developmental gene regulation. We also leveraged these data to link enhancers to putative target genes and demonstrate tissue-specific enrichments of sequence variants associated with disease in humans. The mouse ENCODE data sets provide a compendium of resources for biomedical researchers and achieve, to our knowledge, the most comprehensive view of chromatin dynamics during mammalian fetal development to date.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Datasets as Topic , Fetal Development/genetics , Histones/metabolism , Molecular Sequence Annotation , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/chemistry , Chromatin Immunoprecipitation Sequencing , Disease/genetics , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genetic Variation , Histones/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Organ Specificity/genetics , Reproducibility of Results , Transposases/metabolism
20.
Nucleic Acids Res ; 52(D1): D273-D284, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37956310

ABSTRACT

Although over 170 chemical modifications have been identified, their prevalence, mechanism and function remain largely unknown. To enable integrated analysis of diverse RNA modification profiles, we have developed RMBase v3.0 (http://bioinformaticsscience.cn/rmbase/), a comprehensive platform consisting of eight modules. These modules facilitate the exploration of transcriptome-wide landscape, biogenesis, interactome and functions of RNA modifications. By mining thousands of epitranscriptome datasets with novel pipelines, the 'RNA Modifications' module reveals the map of 73 RNA modifications of 62 species. the 'Genes' module allows to retrieve RNA modification profiles and clusters by gene and transcript. The 'Mechanisms' module explores 23 382 enzyme-catalyzed or snoRNA-guided modified sites to elucidate their biogenesis mechanisms. The 'Co-localization' module systematically formulates potential correlations between 14 histone modifications and 6 RNA modifications in various cell-lines. The 'RMP' module investigates the differential expression profiles of 146 RNA-modifying proteins (RMPs) in 18 types of cancers. The 'Interactome' integrates the interactional relationships between 73 RNA modifications with RBP binding events, miRNA targets and SNPs. The 'Motif' illuminates the enriched motifs for 11 types of RNA modifications identified from epitranscriptome datasets. The 'Tools' introduces a novel web-based 'modGeneTool' for annotating modifications. Overall, RMBase v3.0 provides various resources and tools for studying RNA modifications.


Subject(s)
MicroRNAs , Nucleic Acid Conformation , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Sequence Analysis, RNA , Transcriptome/genetics , Databases, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL