Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Cell ; 182(3): 563-577.e20, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615086

ABSTRACT

Adipose tissues dynamically remodel their cellular composition in response to external cues by stimulating beige adipocyte biogenesis; however, the developmental origin and pathways regulating this process remain insufficiently understood owing to adipose tissue heterogeneity. Here, we employed single-cell RNA-seq and identified a unique subset of adipocyte progenitor cells (APCs) that possessed the cell-intrinsic plasticity to give rise to beige fat. This beige APC population is proliferative and marked by cell-surface proteins, including PDGFRα, Sca1, and CD81. Notably, CD81 is not only a beige APC marker but also required for de novo beige fat biogenesis following cold exposure. CD81 forms a complex with αV/ß1 and αV/ß5 integrins and mediates the activation of integrin-FAK signaling in response to irisin. Importantly, CD81 loss causes diet-induced obesity, insulin resistance, and adipose tissue inflammation. These results suggest that CD81 functions as a key sensor of external inputs and controls beige APC proliferation and whole-body energy homeostasis.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, Beige/metabolism , Energy Metabolism/genetics , Focal Adhesion Kinase 1/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Tetraspanin 28/metabolism , Adipocytes/metabolism , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/growth & development , Adipose Tissue, White/metabolism , Adult , Animals , Ataxin-1/metabolism , Female , Fibronectins/pharmacology , Focal Adhesion Kinase 1/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/genetics , Obesity/metabolism , RNA-Seq , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Single-Cell Analysis , Stem Cells/cytology , Tetraspanin 28/genetics
2.
Cell ; 177(2): 231-242, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951667

ABSTRACT

The Extracellular RNA Communication Consortium (ERCC) was launched to accelerate progress in the new field of extracellular RNA (exRNA) biology and to establish whether exRNAs and their carriers, including extracellular vesicles (EVs), can mediate intercellular communication and be utilized for clinical applications. Phase 1 of the ERCC focused on exRNA/EV biogenesis and function, discovery of exRNA biomarkers, development of exRNA/EV-based therapeutics, and construction of a robust set of reference exRNA profiles for a variety of biofluids. Here, we present progress by ERCC investigators in these areas, and we discuss collaborative projects directed at development of robust methods for EV/exRNA isolation and analysis and tools for sharing and computational analysis of exRNA profiling data.


Subject(s)
Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/metabolism , Extracellular Vesicles/genetics , Biomarkers , Humans , Knowledge Bases , MicroRNAs/genetics , RNA/genetics
3.
Genes Dev ; 36(5-6): 331-347, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35273077

ABSTRACT

Upon fertilization, the mammalian embryo must switch from dependence on maternal transcripts to transcribing its own genome, and in mice this involves the transient up-regulation of MERVL transposons and MERVL-driven genes at the two-cell stage. The mechanisms and requirement for MERVL and two-cell (2C) gene up-regulation are poorly understood. Moreover, this MERVL-driven transcriptional program must be rapidly shut off to allow two-cell exit and developmental progression. Here, we report that robust ribosomal RNA (rRNA) synthesis and nucleolar maturation are essential for exit from the 2C state. 2C-like cells and two-cell embryos show similar immature nucleoli with altered structure and reduced rRNA output. We reveal that nucleolar disruption via blocking RNA polymerase I activity or preventing nucleolar phase separation enhances conversion to a 2C-like state in embryonic stem cells (ESCs) by detachment of the MERVL activator Dux from the nucleolar surface. In embryos, nucleolar disruption prevents proper nucleolar maturation and Dux silencing and leads to two- to four-cell arrest. Our findings reveal an intriguing link between rRNA synthesis, nucleolar maturation, and gene repression during early development.


Subject(s)
Cell Nucleolus , Embryo, Mammalian , Animals , Cell Nucleolus/genetics , Embryonic Development/genetics , Embryonic Stem Cells , Genome , Mammals/genetics , Mice , RNA, Ribosomal/genetics
4.
Cell ; 158(2): 449-461, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036638

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) holds enormous promise for regenerative medicine. To elucidate endogenous barriers limiting this process, we systematically dissected human cellular reprogramming by combining a genome-wide RNAi screen, innovative computational methods, extensive single-hit validation, and mechanistic investigation of relevant pathways and networks. We identify reprogramming barriers, including genes involved in transcription, chromatin regulation, ubiquitination, dephosphorylation, vesicular transport, and cell adhesion. Specific a disintegrin and metalloproteinase (ADAM) proteins inhibit reprogramming, and the disintegrin domain of ADAM29 is necessary and sufficient for this function. Clathrin-mediated endocytosis can be targeted with small molecules and opposes reprogramming by positively regulating TGF-ß signaling. Genetic interaction studies of endocytosis or ubiquitination reveal that barrier pathways can act in linear, parallel, or feedforward loop architectures to antagonize reprogramming. These results provide a global view of barriers to human cellular reprogramming.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , ADAM Proteins/metabolism , Cell Adhesion , Embryonic Stem Cells/metabolism , Endocytosis , Humans , Ubiquitin/metabolism
5.
Genes Dev ; 35(15-16): 1109-1122, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34301766

ABSTRACT

Lung adenocarcinoma, the most prevalent lung cancer subtype, is characterized by its high propensity to metastasize. Despite the importance of metastasis in lung cancer mortality, its underlying cellular and molecular mechanisms remain largely elusive. Here, we identified miR-200 miRNAs as potent suppressors for lung adenocarcinoma metastasis. miR-200 expression is specifically repressed in mouse metastatic lung adenocarcinomas, and miR-200 decrease strongly correlates with poor patient survival. Consistently, deletion of mir-200c/141 in the KrasLSL-G12D/+ ; Trp53flox/flox lung adenocarcinoma mouse model significantly promoted metastasis, generating a desmoplastic tumor stroma highly reminiscent of metastatic human lung cancer. miR-200 deficiency in lung cancer cells promotes the proliferation and activation of adjacent cancer-associated fibroblasts (CAFs), which in turn elevates the metastatic potential of cancer cells. miR-200 regulates the functional interaction between cancer cells and CAFs, at least in part, by targeting Notch ligand Jagged1 and Jagged2 in cancer cells and inducing Notch activation in adjacent CAFs. Hence, the interaction between cancer cells and CAFs constitutes an essential mechanism to promote metastatic potential.


Subject(s)
Cancer-Associated Fibroblasts , Lung Neoplasms , MicroRNAs , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis/pathology
6.
Cell ; 152(4): 909-22, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23394947

ABSTRACT

Genetic interaction (GI) maps, comprising pairwise measures of how strongly the function of one gene depends on the presence of a second, have enabled the systematic exploration of gene function in microorganisms. Here, we present a two-stage strategy to construct high-density GI maps in mammalian cells. First, we use ultracomplex pooled shRNA libraries (25 shRNAs/gene) to identify high-confidence hit genes for a given phenotype and effective shRNAs. We then construct double-shRNA libraries from these to systematically measure GIs between hits. A GI map focused on ricin susceptibility broadly recapitulates known pathways and provides many unexpected insights. These include a noncanonical role for COPI, a previously uncharacterized protein complex affecting toxin clearance, a specialized role for the ribosomal protein RPS25, and functionally distinct mammalian TRAPP complexes. The ability to rapidly generate mammalian GI maps provides a potentially transformative tool for defining gene function and designing combination therapies based on synergistic pairs.


Subject(s)
Biological Transport , Epistasis, Genetic , Ricin/toxicity , Atorvastatin , Carrier Proteins/metabolism , Cell Line, Tumor , Coat Protein Complex I/metabolism , Endoplasmic Reticulum/metabolism , Heptanoic Acids/pharmacology , Humans , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Pyrroles/pharmacology , RNA, Small Interfering , Ribosomal Proteins/metabolism , Vesicular Transport Proteins/metabolism
7.
Nat Rev Genet ; 22(7): 448-458, 2021 07.
Article in English | MEDLINE | ID: mdl-33824487

ABSTRACT

The presence of RNAs in the extracellular milieu has sparked the hypothesis that RNA may play a role in mammalian cell-cell communication. As functional nucleic acids transfer from cell to cell in plants and nematodes, the idea that mammalian cells also transfer functional extracellular RNA (exRNA) is enticing. However, untangling the role of mammalian exRNAs poses considerable experimental challenges. This Review discusses the evidence for and against functional exRNAs in mammals and their proposed roles in health and disease, such as cancer and cardiovascular disease. We conclude with a discussion of the forward-looking prospects for studying the potential of mammalian exRNAs as mediators of cell-cell communication.


Subject(s)
Mammals/genetics , RNA/physiology , Animals , Extracellular Space/physiology , Humans , Mammals/physiology
8.
Nat Immunol ; 15(4): 393-401, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24608041

ABSTRACT

The microRNA miR-210 is a signature of hypoxia. We found robust increase in the abundance of miR-210 (>100-fold) in activated T cells, especially in the TH17 lineage of helper T cells. Hypoxia acted in synergy with stimulation via the T cell antigen receptor (TCR) and coreceptor CD28 to accelerate and increase Mir210 expression. Mir210 was directly regulated by HIF-1α, a key transcriptional regulator of TH17 polarization. Unexpectedly, we identified Hif1a as a target of miR-210, which suggested negative feedback by miR-210 in inhibiting HIF-1α expression. Deletion of Mir210 promoted TH17 differentiation under conditions of limited oxygen. In experimental colitis, miR-210 reduced the abundance of Hif1a transcripts and the proportion of cells that produced inflammatory cytokines and controlled disease severity. Our study identifies miR-210 as an important regulator of T cell differentiation in hypoxia, which can limit immunopathology.


Subject(s)
Colitis, Ulcerative/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/metabolism , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Animals , CD4 Antigens/metabolism , Cell Differentiation/genetics , Cell Hypoxia/immunology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , RNA Interference/immunology , T-Lymphocyte Subsets/cytology , Th17 Cells/cytology
9.
Mol Cell ; 69(2): 265-278.e6, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351846

ABSTRACT

While Slicer activity of Argonaute is central to RNAi, conserved roles of slicing in endogenous regulatory biology are less clear, especially in mammals. Biogenesis of erythroid Dicer-independent mir-451 involves Ago2 catalysis, but mir-451-KO mice do not phenocopy Ago2 catalytic-dead (Ago2-CD) mice, suggesting other needs for slicing. Here, we reveal mir-486 as another dominant erythroid miRNA with atypical biogenesis. While it is Dicer dependent, it requires slicing to eliminate its star strand. Thus, in Ago2-CD conditions, miR-486-5p is functionally inactive due to duplex arrest. Genome-wide analyses reveal miR-486 and miR-451 as the major slicing-dependent miRNAs in the hematopoietic system. Moreover, mir-486-KO mice exhibit erythroid defects, and double knockout of mir-486/451 phenocopies the cell-autonomous effects of Ago2-CD in the hematopoietic system. Finally, we observe that Ago2 is the dominant-expressed Argonaute in maturing erythroblasts, reflecting a specialized environment for processing slicing-dependent miRNAs. Overall, the mammalian hematopoietic system has evolved multiple conserved requirements for Slicer-dependent miRNA biogenesis.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/genetics , Animals , Argonaute Proteins/genetics , Argonaute Proteins/physiology , DEAD-box RNA Helicases/metabolism , Erythroblasts/metabolism , Genome-Wide Association Study , Mammals/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , RNA Interference , Ribonuclease III/metabolism , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid
10.
Immunity ; 44(4): 821-32, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26850657

ABSTRACT

MicroRNAs (miRNAs) are important regulators of cell fate decisions in immune responses. They act by coordinate repression of multiple target genes, a property that we exploited to uncover regulatory networks that govern T helper-2 (Th2) cells. A functional screen of individual miRNAs in primary T cells uncovered multiple miRNAs that inhibited Th2 cell differentiation. Among these were miR-24 and miR-27, miRNAs coexpressed from two genomic clusters, which each functioned independently to limit interleukin-4 (IL-4) production. Mice lacking both clusters in T cells displayed increased Th2 cell responses and tissue pathology in a mouse model of asthma. Gene expression and pathway analyses placed miR-27 upstream of genes known to regulate Th2 cells. They also identified targets not previously associated with Th2 cell biology which regulated IL-4 production in unbiased functional testing. Thus, elucidating the biological function and target repertoire of miR-24 and miR-27 reveals regulators of Th2 cell biology.


Subject(s)
Asthma/immunology , Interleukin-4/biosynthesis , MicroRNAs/genetics , Th2 Cells/immunology , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Female , Inflammation/immunology , Interleukin-4/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Multigene Family/genetics , Sequence Analysis, RNA , Th2 Cells/cytology
11.
Nature ; 572(7771): 614-619, 2019 08.
Article in English | MEDLINE | ID: mdl-31435015

ABSTRACT

Branched-chain amino acid (BCAA; valine, leucine and isoleucine) supplementation is often beneficial to energy expenditure; however, increased circulating levels of BCAA are linked to obesity and diabetes. The mechanisms of this paradox remain unclear. Here we report that, on cold exposure, brown adipose tissue (BAT) actively utilizes BCAA in the mitochondria for thermogenesis and promotes systemic BCAA clearance in mice and humans. In turn, a BAT-specific defect in BCAA catabolism attenuates systemic BCAA clearance, BAT fuel oxidation and thermogenesis, leading to diet-induced obesity and glucose intolerance. Mechanistically, active BCAA catabolism in BAT is mediated by SLC25A44, which transports BCAAs into mitochondria. Our results suggest that BAT serves as a key metabolic filter that controls BCAA clearance via SLC25A44, thereby contributing to the improvement of metabolic health.


Subject(s)
Adipose Tissue, Brown/metabolism , Amino Acid Transport Systems/metabolism , Amino Acids, Branched-Chain/metabolism , Energy Metabolism , Homeostasis , Mitochondrial Proteins/metabolism , Solute Carrier Proteins/metabolism , Thermogenesis , Adipose Tissue, Brown/cytology , Animals , Cold Temperature , Glucose Intolerance/metabolism , Humans , Male , Mice , Mitochondria/metabolism , Obesity/metabolism
12.
Stem Cells ; 41(5): 520-539, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36945732

ABSTRACT

Epigenetic mechanisms regulate the multilineage differentiation capacity of hematopoietic stem cells (HSCs) into a variety of blood and immune cells. Mapping the chromatin dynamics of functionally defined cell populations will shed mechanistic insight into 2 major, unanswered questions in stem cell biology: how does epigenetic identity contribute to a cell type's lineage potential, and how do cascades of chromatin remodeling dictate ensuing fate decisions? Our recent work revealed evidence of multilineage gene priming in HSCs, where open cis-regulatory elements (CREs) exclusively shared between HSCs and unipotent lineage cells were enriched for DNA binding motifs of known lineage-specific transcription factors. Oligopotent progenitor populations operating between the HSCs and unipotent cells play essential roles in effecting hematopoietic homeostasis. To test the hypothesis that selective HSC-primed lineage-specific CREs remain accessible throughout differentiation, we used ATAC-seq to map the temporal dynamics of chromatin remodeling during progenitor differentiation. We observed epigenetic-driven clustering of oligopotent and unipotent progenitors into distinct erythromyeloid and lymphoid branches, with multipotent HSCs and MPPs associating with the erythromyeloid lineage. We mapped the dynamics of lineage-primed CREs throughout hematopoiesis and identified both unique and shared CREs as potential lineage reinforcement mechanisms at fate branch points. Additionally, quantification of genome-wide peak count and size revealed overall greater chromatin accessibility in HSCs, allowing us to identify HSC-unique peaks as putative regulators of self-renewal and multilineage potential. Finally, CRISPRi-mediated targeting of ATACseq-identified putative CREs in HSCs allowed us to demonstrate the functional role of selective CREs in lineage-specific gene expression. These findings provide insight into the regulation of stem cell multipotency and lineage commitment throughout hematopoiesis and serve as a resource to test functional drivers of hematopoietic lineage fate.


Subject(s)
Chromatin , Hematopoiesis , Chromatin/genetics , Chromatin/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics
13.
Nature ; 551(7679): 247-250, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29088702

ABSTRACT

Acquired drug resistance prevents cancer therapies from achieving stable and complete responses. Emerging evidence implicates a key role for non-mutational drug resistance mechanisms underlying the survival of residual cancer 'persister' cells. The persister cell pool constitutes a reservoir from which drug-resistant tumours may emerge. Targeting persister cells therefore presents a therapeutic opportunity to impede tumour relapse. We previously found that cancer cells in a high mesenchymal therapy-resistant cell state are dependent on the lipid hydroperoxidase GPX4 for survival. Here we show that a similar therapy-resistant cell state underlies the behaviour of persister cells derived from a wide range of cancers and drug treatments. Consequently, we demonstrate that persister cells acquire a dependency on GPX4. Loss of GPX4 function results in selective persister cell ferroptotic death in vitro and prevents tumour relapse in mice. These findings suggest that targeting of GPX4 may represent a therapeutic strategy to prevent acquired drug resistance.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Glutathione Peroxidase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Antioxidants/metabolism , Drug Evaluation, Preclinical , Female , Humans , Iron/metabolism , Male , Mesoderm/drug effects , Mesoderm/enzymology , Mesoderm/pathology , Mice , Molecular Targeted Therapy , Neoplasms/enzymology , Phospholipid Hydroperoxide Glutathione Peroxidase , Recurrence , Xenograft Model Antitumor Assays
14.
Mol Cell ; 58(4): 575-85, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000843

ABSTRACT

The most widely used approach for defining gene function is to reduce or completely disrupt its normal expression. For over a decade, RNAi has ruled the lab, offering a magic bullet to disrupt gene expression in many organisms. However, new biotechnological tools--specifically CRISPR-based technologies--have become available and are squeezing out RNAi dominance in mammalian cell studies. These seemingly competing technologies leave research investigators with the question: "Which technology should I use in my experiment?" This review offers a practical resource to compare and contrast these technologies, guiding the investigator when and where to use this fantastic array of powerful tools.


Subject(s)
CRISPR-Cas Systems/genetics , Deoxyribonucleases/metabolism , Genetic Engineering/methods , RNA Interference , Animals , Biomedical Research/methods , Biomedical Research/trends , Biotechnology/methods , Biotechnology/trends , Genetic Engineering/trends , Humans , Models, Genetic
15.
PLoS Biol ; 16(12): e3000067, 2018 12.
Article in English | MEDLINE | ID: mdl-30532236

ABSTRACT

This Formal Comment responds to a recent Meta-Research Article by identifying initiatives that are already in place for funding risky exploratory research that illuminate mysteries of the dark genome.


Subject(s)
Genome , Research
16.
Proc Natl Acad Sci U S A ; 115(17): E4051-E4060, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29632189

ABSTRACT

Despite decades of research, mechanisms controlling T cell activation remain only partially understood, which hampers T cell-based immune cancer therapies. Here, we performed a genome-wide CRISPR screen to search for genes that regulate T cell activation. Our screen confirmed many of the known regulators in proximal T cell receptor signaling and, importantly, also uncovered a previously uncharacterized regulator, FAM49B (family with sequence similarity 49 member B). FAM49B deficiency led to hyperactivation of Jurkat T cells following T cell receptor stimulation, as indicated by enhancement of CD69 induction, PAK phosphorylation, and actin assembly. FAM49B directly interacted with the active form of the small GTPase Rac, and genetic disruption of the FAM49B-Rac interaction compromised FAM49B function. Thus, FAM49B inhibits T cell activation by repressing Rac activity and modulating cytoskeleton reorganization.


Subject(s)
Lymphocyte Activation , Neoplasm Proteins/immunology , T-Lymphocytes/immunology , Actins/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CRISPR-Cas Systems , Cytoskeleton/genetics , Cytoskeleton/immunology , Genome-Wide Association Study , Humans , Jurkat Cells , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , T-Lymphocytes/cytology
17.
Genome Res ; 27(1): 38-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27831498

ABSTRACT

Candidate enhancers can be identified on the basis of chromatin modifications, the binding of chromatin modifiers and transcription factors and cofactors, or chromatin accessibility. However, validating such candidates as bona fide enhancers requires functional characterization, typically achieved through reporter assays that test whether a sequence can increase expression of a transcriptional reporter via a minimal promoter. A longstanding concern is that reporter assays are mainly implemented on episomes, which are thought to lack physiological chromatin. However, the magnitude and determinants of differences in cis-regulation for regulatory sequences residing in episomes versus chromosomes remain almost completely unknown. To address this systematically, we developed and applied a novel lentivirus-based massively parallel reporter assay (lentiMPRA) to directly compare the functional activities of 2236 candidate liver enhancers in an episomal versus a chromosomally integrated context. We find that the activities of chromosomally integrated sequences are substantially different from the activities of the identical sequences assayed on episomes, and furthermore are correlated with different subsets of ENCODE annotations. The results of chromosomally based reporter assays are also more reproducible and more strongly predictable by both ENCODE annotations and sequence-based models. With a linear model that combines chromatin annotations and sequence information, we achieve a Pearson's R2 of 0.362 for predicting the results of chromosomally integrated reporter assays. This level of prediction is better than with either chromatin annotations or sequence information alone and also outperforms predictive models of episomal assays. Our results have broad implications for how cis-regulatory elements are identified, prioritized and functionally validated.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Plasmids/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomes/genetics , Genes, Reporter , High-Throughput Nucleotide Sequencing , Humans , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors
18.
Development ; 144(13): 2517-2528, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28576768

ABSTRACT

The tear-producing lacrimal gland is a tubular organ that protects and lubricates the ocular surface. The lacrimal gland possesses many features that make it an excellent model in which to investigate tubulogenesis, but the cell types and lineage relationships that drive lacrimal gland formation are unclear. Using single-cell sequencing and other molecular tools, we reveal novel cell identities and epithelial lineage dynamics that underlie lacrimal gland development. We show that the lacrimal gland from its earliest developmental stages is composed of multiple subpopulations of immune, epithelial and mesenchymal cell lineages. The epithelial lineage exhibits the most substantial cellular changes, transitioning through a series of unique transcriptional states to become terminally differentiated acinar, ductal and myoepithelial cells. Furthermore, lineage tracing in postnatal and adult glands provides the first direct evidence of unipotent KRT5+ epithelial cells in the lacrimal gland. Finally, we show conservation of developmental markers between the developing mouse and human lacrimal gland, supporting the use of mice to understand human development. Together, our data reveal crucial features of lacrimal gland development that have broad implications for understanding epithelial organogenesis.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Lacrimal Apparatus/cytology , Lacrimal Apparatus/embryology , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Biomarkers/metabolism , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Mice , Phenotype , Sequence Analysis, RNA , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism
19.
Immunity ; 34(1): 50-60, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21236706

ABSTRACT

Self-reactive T cell clones that escape negative selection are either deleted or rendered functionally unresponsive (anergic), thus preventing them from propagating host tissue damage. By using an in vivo model, we investigated molecular mechanisms for T cell tolerance, finding that despite a characteristic inability to generate effector cytokine proteins, self-reactive T cells express large amounts of cytokine mRNAs. This disconnect between cytokine message and protein was not observed in T cells mounting productive responses to foreign antigens but, instead, was seen only in those responding to self, where the block in protein translation was shown to involve conserved AU-rich elements within cytokine 3'UTRs. These studies reveal that translation of abundant cytokine mRNAs is limited in self-reactive T cells and, thus, identify posttranscriptional silencing of antigen-driven gene expression as a key mechanism underlying the anergic phenotype of self-reactive T cells.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , 3' Untranslated Regions/genetics , Adoptive Transfer , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Clonal Anergy , Cytokines/genetics , Cytokines/immunology , Immune Tolerance , Mice , Mice, Transgenic , Protein Biosynthesis/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA-Induced Silencing Complex/immunology , Response Elements/genetics
20.
Mol Cell ; 46(4): 507-17, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22503104

ABSTRACT

Assembly of microRNA ribonucleoproteins (miRNPs) or RNA-induced silencing complexes (RISCs) is essential for the function of miRNAs and initiates from processing of precursor miRNAs (pre-miRNAs) by Dicer or by Ago2. Here, we report an in vitro miRNP/RISC assembly assay programmed by pre-miRNAs from mammalian cell lysates. Combining in vivo studies in Dicer Knockout cells reconstituted with wild-type or catalytically inactive Dicer, we find that the miRNA loading complex (miRLC) is the primary machinery linking pre-miRNA processing to miRNA loading. We show that a miRNA precursor deposit complex (miPDC) plays a crucial role in Dicer-independent miRNA biogenesis and promotes miRNP assembly of certain Dicer-dependent miRNAs. Furthermore, we find that 5'-uridine, 3'-mid base pairing, and 5'-mid mismatches within pre-miRNAs promote their assembly into miPDC. Our studies provide a comprehensive view of miRNP/RISC assembly pathways in mammals, and our assay provides a versatile platform for further mechanistic dissection of such pathways in mammals.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Cell Line , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , In Vitro Techniques , Mice , Mice, Knockout , MicroRNAs/chemistry , Models, Biological , Protein Multimerization , RNA Processing, Post-Transcriptional , RNA-Induced Silencing Complex/chemistry , Ribonuclease III/chemistry , Ribonuclease III/deficiency , Ribonuclease III/genetics , Ribonuclease III/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL