Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 38(8): 954-961, 2020 08.
Article in English | MEDLINE | ID: mdl-32231336

ABSTRACT

Single-cell CRISPR screens enable the exploration of mammalian gene function and genetic regulatory networks. However, use of this technology has been limited by reliance on indirect indexing of single-guide RNAs (sgRNAs). Here we present direct-capture Perturb-seq, a versatile screening approach in which expressed sgRNAs are sequenced alongside single-cell transcriptomes. Direct-capture Perturb-seq enables detection of multiple distinct sgRNA sequences from individual cells and thus allows pooled single-cell CRISPR screens to be easily paired with combinatorial perturbation libraries that contain dual-guide expression vectors. We demonstrate the utility of this approach for high-throughput investigations of genetic interactions and, leveraging this ability, dissect epistatic interactions between cholesterol biogenesis and DNA repair. Using direct capture Perturb-seq, we also show that targeting individual genes with multiple sgRNAs per cell improves efficacy of CRISPR interference and activation, facilitating the use of compact, highly active CRISPR libraries for single-cell screens. Last, we show that hybridization-based target enrichment permits sensitive, specific sequencing of informative transcripts from single-cell RNA-seq experiments.


Subject(s)
CRISPR-Cas Systems , Nucleic Acid Amplification Techniques/methods , RNA, Guide, Kinetoplastida/genetics , Gene Expression Regulation , Gene Targeting , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Single-Cell Analysis , Transcriptome
2.
Cell ; 179(2): 403-416.e23, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585080

ABSTRACT

Pulmonary neuroendocrine (NE) cells are neurosensory cells sparsely distributed throughout the bronchial epithelium, many in innervated clusters of 20-30 cells. Following lung injury, NE cells proliferate and generate other cell types to promote epithelial repair. Here, we show that only rare NE cells, typically 2-4 per cluster, function as stem cells. These fully differentiated cells display features of classical stem cells. Most proliferate (self-renew) following injury, and some migrate into the injured area. A week later, individual cells, often just one per cluster, lose NE identity (deprogram), transit amplify, and reprogram to other fates, creating large clonal repair patches. Small cell lung cancer (SCLC) tumor suppressors regulate the stem cells: Rb and p53 suppress self-renewal, whereas Notch marks the stem cells and initiates deprogramming and transit amplification. We propose that NE stem cells give rise to SCLC, and transformation results from constitutive activation of stem cell renewal and inhibition of deprogramming.


Subject(s)
Cell Transformation, Neoplastic/pathology , Lung Neoplasms/pathology , Lung/pathology , Neoplastic Stem Cells/pathology , Neuroendocrine Cells/pathology , Receptors, Notch/metabolism , Retinoblastoma Protein/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Lung Injury/pathology , Lung Neoplasms/metabolism , Mice , Neoplastic Stem Cells/metabolism , Neuroendocrine Cells/metabolism , Single-Cell Analysis/methods , Small Cell Lung Carcinoma/metabolism
3.
Elife ; 72018 01 10.
Article in English | MEDLINE | ID: mdl-29319504

ABSTRACT

A major challenge in biology is identifying distinct cell classes and mapping their interactions in vivo. Tissue-dissociative technologies enable deep single cell molecular profiling but do not provide spatial information. We developed a proximity ligation in situ hybridization technology (PLISH) with exceptional signal strength, specificity, and sensitivity in tissue. Multiplexed data sets can be acquired using barcoded probes and rapid label-image-erase cycles, with automated calculation of single cell profiles, enabling clustering and anatomical re-mapping of cells. We apply PLISH to expression profile ~2900 cells in intact mouse lung, which identifies and localizes known cell types, including rare ones. Unsupervised classification of the cells indicates differential expression of 'housekeeping' genes between cell types, and re-mapping of two sub-classes of Club cells highlights their segregated spatial domains in terminal airways. By enabling single cell profiling of various RNA species in situ, PLISH can impact many areas of basic and medical research.


Subject(s)
Automation, Laboratory/methods , Cells/classification , DNA Fingerprinting/methods , In Situ Hybridization/methods , Pathology, Molecular/methods , Single-Cell Analysis/methods , Animals , Lung/cytology , Mice, Inbred C57BL , Sensitivity and Specificity
4.
Nat Chem Biol ; 14(1): 15-21, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29106397

ABSTRACT

In principle, the millisecond emission lifetimes of lanthanide chelates should enable their ultrasensitive detection in biological systems by time-resolved optical microscopy. In practice, however, lanthanide imaging techniques have provided no better sensitivity than conventional fluorescence microscopy. Here, we identified three fundamental problems that have impeded lanthanide microscopy: low photon flux, inefficient excitation, and optics-derived background luminescence. We overcame these limitations with a new lanthanide imaging modality, transreflected illumination with luminescence resonance energy transfer (trLRET), which increases the time-integrated signal intensities of lanthanide lumiphores by 170-fold and the signal-to-background ratios by 75-fold. We demonstrate that trLRET provides at least an order-of-magnitude increase in detection sensitivity over that of conventional epifluorescence microscopy when used to visualize endogenous protein expression in zebrafish embryos. We also show that trLRET can be used to optically detect molecular interactions in vivo. trLRET promises to unlock the full potential of lanthanide lumiphores for ultrasensitive, autofluorescence-free biological imaging.


Subject(s)
Coordination Complexes/chemistry , Lanthanoid Series Elements/chemistry , Luminescent Agents/chemistry , Luminescent Measurements/methods , Optical Imaging/methods , Zebrafish Proteins/biosynthesis , Animals , Coordination Complexes/chemical synthesis , Lanthanoid Series Elements/chemical synthesis , Luminescent Agents/chemical synthesis , Sensitivity and Specificity , Zebrafish/embryology , Zebrafish/metabolism
5.
Mol Pharmacol ; 91(2): 145-156, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27879340

ABSTRACT

The ability of chemically distinct ligands to produce different effects on the same G protein-coupled receptor (GPCR) has interesting therapeutic implications, but, if excessively propagated downstream, would introduce biologic noise compromising cognate ligand detection. We asked whether cells have the ability to limit the degree to which chemical diversity imposed at the ligand-GPCR interface is propagated to the downstream signal. We carried out an unbiased analysis of the integrated cellular response elicited by two chemically and pharmacodynamically diverse ß-adrenoceptor agonists, isoproterenol and salmeterol. We show that both ligands generate an identical integrated response, and that this stereotyped output requires endocytosis. We further demonstrate that the endosomal ß2-adrenergic receptor signal confers uniformity on the downstream response because it is highly sensitive and saturable. Based on these findings, we propose that GPCR signaling from endosomes functions as a biologic noise filter to enhance reliability of cognate ligand detection.


Subject(s)
Endocytosis , Receptors, G-Protein-Coupled/metabolism , Endosomes/drug effects , Endosomes/metabolism , HEK293 Cells , Humans , Isoproterenol/pharmacology , Ligands , Mass Spectrometry , Models, Biological , Oligonucleotide Array Sequence Analysis , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proteome/metabolism , Proteomics , Receptors, Adrenergic, beta-2/metabolism , Salmeterol Xinafoate/pharmacology , Signal Transduction/drug effects , Transcription, Genetic/drug effects
6.
Elife ; 42015 Oct 19.
Article in English | MEDLINE | ID: mdl-26479710

ABSTRACT

Epicardial cells on the heart's surface give rise to coronary artery smooth muscle cells (caSMCs) located deep in the myocardium. However, the differentiation steps between epicardial cells and caSMCs are unknown as are the final maturation signals at coronary arteries. Here, we use clonal analysis and lineage tracing to show that caSMCs derive from pericytes, mural cells associated with microvessels, and that these cells are present in adults. During development following the onset of blood flow, pericytes at arterial remodeling sites upregulate Notch3 while endothelial cells express Jagged-1. Deletion of Notch3 disrupts caSMC differentiation. Our data support a model wherein epicardial-derived pericytes populate the entire coronary microvasculature, but differentiate into caSMCs at arterial remodeling zones in response to Notch signaling. Our data are the first demonstration that pericytes are progenitors for smooth muscle, and their presence in adult hearts reveals a new potential cell type for targeting during cardiovascular disease.


Subject(s)
Cell Differentiation , Coronary Vessels/cytology , Muscle Cells/physiology , Muscle, Smooth/cytology , Pericytes/physiology , Stem Cells/physiology , Animals , Mice, Inbred C57BL , Receptor, Notch3 , Receptors, Notch/biosynthesis , Up-Regulation
7.
PLoS One ; 10(7): e0128975, 2015.
Article in English | MEDLINE | ID: mdl-26176839

ABSTRACT

Characterization of the molecular attributes and spatial arrangements of cells and features within complex human tissues provides a critical basis for understanding processes involved in development and disease. Moreover, the ability to automate steps in the analysis and interpretation of histological images that currently require manual inspection by pathologists could revolutionize medical diagnostics. Toward this end, we developed a new imaging approach called multidimensional microscopic molecular profiling (MMMP) that can measure several independent molecular properties in situ at subcellular resolution for the same tissue specimen. MMMP involves repeated cycles of antibody or histochemical staining, imaging, and signal removal, which ultimately can generate information analogous to a multidimensional flow cytometry analysis on intact tissue sections. We performed a MMMP analysis on a tissue microarray containing a diverse set of 102 human tissues using a panel of 15 informative antibody and 5 histochemical stains plus DAPI. Large-scale unsupervised analysis of MMMP data, and visualization of the resulting classifications, identified molecular profiles that were associated with functional tissue features. We then directly annotated H&E images from this MMMP series such that canonical histological features of interest (e.g. blood vessels, epithelium, red blood cells) were individually labeled. By integrating image annotation data, we identified molecular signatures that were associated with specific histological annotations and we developed statistical models for automatically classifying these features. The classification accuracy for automated histology labeling was objectively evaluated using a cross-validation strategy, and significant accuracy (with a median per-pixel rate of 77% per feature from 15 annotated samples) for de novo feature prediction was obtained. These results suggest that high-dimensional profiling may advance the development of computer-based systems for automatically parsing relevant histological and cellular features from molecular imaging data of arbitrary human tissue samples, and can provide a framework and resource to spur the optimization of these technologies.


Subject(s)
Algorithms , Image Processing, Computer-Assisted/methods , Microscopy/methods , Molecular Imaging/methods , Automation , Humans , Software , Tissue Array Analysis , Unsupervised Machine Learning
8.
PLoS One ; 9(10): e110799, 2014.
Article in English | MEDLINE | ID: mdl-25353621

ABSTRACT

We developed a novel technique, called pseudouridine site identification sequencing (PSI-seq), for the transcriptome-wide mapping of pseudouridylation sites with single-base resolution from cellular RNAs based on the induced termination of reverse transcription specifically at pseudouridines following CMCT treatment. PSI-seq analysis of RNA samples from S. cerevisiae correctly detected all of the 43 known pseudouridines in yeast 18S and 25S ribosomal RNA with high specificity. Moreover, application of PSI-seq to the yeast transcriptome revealed the presence of site-specific pseudouridylation within dozens of mRNAs, including RPL11a, TEF1, and other genes implicated in translation. To identify the mechanisms responsible for mRNA pseudouridylation, we genetically deleted candidate pseudouridine synthase (Pus) enzymes and reconstituted their activities in vitro. These experiments demonstrated that the Pus1 enzyme was necessary and sufficient for pseudouridylation of RPL11a mRNA, whereas Pus4 modified TEF1 mRNA, and Pus6 pseudouridylated KAR2 mRNA. Finally, we determined that modification of RPL11a at Ψ -68 was observed in RNA from the related yeast S. mikitae, and Ψ -239 in TEF1 mRNA was maintained in S. mikitae as well as S. pombe, indicating that these pseudouridylations are ancient, evolutionarily conserved RNA modifications. This work establishes that site-specific pseudouridylation of eukaryotic mRNAs is a genetically programmed RNA modification that naturally occurs in multiple yeast transcripts via distinct mechanisms, suggesting that mRNA pseudouridylation may provide an important novel regulatory function. The approach and strategies that we report here should be generally applicable to the discovery of pseudouridylation, or other RNA modifications, in diverse biological contexts.


Subject(s)
Intramolecular Transferases/metabolism , Pseudouridine/analysis , RNA, Messenger/chemistry , Saccharomyces cerevisiae/genetics , Sequence Analysis, RNA/methods , Gene Expression Profiling/methods , Intramolecular Transferases/genetics , RNA Processing, Post-Transcriptional , RNA, Fungal/chemistry , RNA, Fungal/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
9.
PLoS Genet ; 8(7): e1002862, 2012.
Article in English | MEDLINE | ID: mdl-22844259

ABSTRACT

The unfolded protein response (UPR) is a conserved mechanism that mitigates accumulation of unfolded proteins in the ER. The yeast UPR is subject to intricate post-transcriptional regulation, involving recruitment of the RNA encoding the Hac1 transcription factor to the ER and its unconventional splicing. To investigate the mechanisms underlying regulation of the UPR, we screened the yeast proteome for proteins that specifically interact with HAC1 RNA. Protein microarray experiments revealed that HAC1 interacts specifically with small ras GTPases of the Ypt family. We characterized the interaction of HAC1 RNA with one of these proteins, the yeast Rab1 homolog Ypt1. We found that Ypt1 protein specifically associated in vivo with unspliced HAC1 RNA. This association was disrupted by conditions that impaired protein folding in the ER and induced the UPR. Also, the Ypt1-HAC1 interaction depended on IRE1 and ADA5, the two genes critical for UPR activation. Decreasing expression of the Ypt1 protein resulted in a reduced rate of HAC1 RNA decay, leading to significantly increased levels of both unspliced and spliced HAC1 RNA, and delayed attenuation of the UPR, when ER stress was relieved. Our findings establish that Ypt1 contributes to regulation of UPR signaling dynamics by promoting the decay of HAC1 RNA, suggesting a potential regulatory mechanism for linking vesicle trafficking to the UPR and ER homeostasis.


Subject(s)
Basic-Leucine Zipper Transcription Factors , RNA Stability/genetics , Repressor Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Unfolded Protein Response/genetics , rab GTP-Binding Proteins , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Fungal , Protein Array Analysis , Protein Folding , Protein Interaction Maps , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
10.
Nucleic Acids Res ; 39(4): 1501-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20959291

ABSTRACT

Post-transcriptional regulation of gene expression, including mRNA localization, translation and decay, is ubiquitous yet still largely unexplored. How is the post-transcriptional regulatory program of each mRNA encoded in its sequence? Hundreds of specific RNA-binding proteins (RBPs) appear to play roles in mediating the post-transcriptional regulatory program, akin to the roles of specific DNA-binding proteins in transcription. As a step toward decoding the regulatory programs encoded in each mRNA, we focused on specific mRNA-protein interactions. We computationally analyzed the sequences of Saccharomyces cerevisiae mRNAs bound in vivo by 29 specific RBPs, identifying eight novel candidate motifs and confirming or extending six earlier reported recognition elements. Biochemical selections for RNA sequences selectively recognized by 12 yeast RBPs yielded novel motifs bound by Pin4, Nsr1, Hrb1, Gbp2, Sgn1 and Mrn1, and recovered the known recognition elements for Puf3, She2, Vts1 and Whi3. Most of the RNA elements we uncovered were associated with coherent mRNA expression changes and were significantly conserved in related yeasts, supporting their functional importance and suggesting that the corresponding RNA-protein interactions are evolutionarily conserved.


Subject(s)
RNA, Fungal/chemistry , RNA, Messenger/chemistry , Regulatory Sequences, Ribonucleic Acid , Saccharomyces cerevisiae/genetics , Binding Sites , Computational Biology/methods , Gene Expression Profiling , Phylogeny , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , SELEX Aptamer Technique , Saccharomyces cerevisiae/metabolism
11.
PLoS Biol ; 6(10): e255, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18959479

ABSTRACT

RNA-binding proteins (RBPs) have roles in the regulation of many post-transcriptional steps in gene expression, but relatively few RBPs have been systematically studied. We searched for the RNA targets of 40 proteins in the yeast Saccharomyces cerevisiae: a selective sample of the approximately 600 annotated and predicted RBPs, as well as several proteins not annotated as RBPs. At least 33 of these 40 proteins, including three of the four proteins that were not previously known or predicted to be RBPs, were reproducibly associated with specific sets of a few to several hundred RNAs. Remarkably, many of the RBPs we studied bound mRNAs whose protein products share identifiable functional or cytotopic features. We identified specific sequences or predicted structures significantly enriched in target mRNAs of 16 RBPs. These potential RNA-recognition elements were diverse in sequence, structure, and location: some were found predominantly in 3'-untranslated regions, others in 5'-untranslated regions, some in coding sequences, and many in two or more of these features. Although this study only examined a small fraction of the universe of yeast RBPs, 70% of the mRNA transcriptome had significant associations with at least one of these RBPs, and on average, each distinct yeast mRNA interacted with three of the RBPs, suggesting the potential for a rich, multidimensional network of regulation. These results strongly suggest that combinatorial binding of RBPs to specific recognition elements in mRNAs is a pervasive mechanism for multi-dimensional regulation of their post-transcriptional fate.


Subject(s)
RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Databases, Protein , Oligonucleotide Array Sequence Analysis , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Untranslated Regions/genetics , Untranslated Regions/metabolism
12.
Genome Res ; 16(12): 1505-16, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17038564

ABSTRACT

Nucleosome positions within the chromatin landscape are known to serve as a major determinant of DNA accessibility to transcription factors and other interacting components. To delineate nucleosomal patterns in a model genetic organism, Caenorhabditis elegans, we have carried out a genome-wide analysis in which DNA fragments corresponding to nucleosome cores were liberated using an enzyme (micrococcal nuclease) with a strong preference for cleavage in non-nucleosomal regions. Sequence analysis of 284,091 putative nucleosome cores obtained in this manner from a mixed-stage population of C. elegans reveals a combined picture of flexibility and constraint in nucleosome positioning. As has previously been observed in studies of individual loci in diverse biological systems, we observe areas in the genome where nucleosomes can adopt a wide variety of positions in a given region, areas with little or no nucleosome coverage, and areas where nucleosomes reproducibly adopt a specific positional pattern. In addition to illuminating numerous aspects of chromatin structure for C. elegans, this analysis provides a reference from which to begin an investigation of relationships between the nucleosomal pattern, chromosomal architecture, and lineage-based gene activity on a genome-wide scale.


Subject(s)
Caenorhabditis elegans/genetics , Chromatin/chemistry , Nucleosomes/chemistry , Amino Acid Sequence , Animals , Chromatin/genetics , Chromatin/metabolism , DNA, Helminth/analysis , Genome, Helminth , Micrococcal Nuclease/pharmacology , Molecular Sequence Data , Nucleosomes/genetics , Sequence Analysis, DNA , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...