scholarly journals CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells

Science ◽  
2016 ◽  
Vol 355 (6320) ◽  
pp. eaah7111 ◽  
Author(s):  
S. John Liu ◽  
Max A. Horlbeck ◽  
Seung Woo Cho ◽  
Harjus S. Birk ◽  
Martina Malatesta ◽  
...  

The human genome produces thousands of long noncoding RNAs (lncRNAs)—transcripts >200 nucleotides long that do not encode proteins. Although critical roles in normal biology and disease have been revealed for a subset of lncRNAs, the function of the vast majority remains untested. We developed a CRISPR interference (CRISPRi) platform targeting 16,401 lncRNA loci in seven diverse cell lines, including six transformed cell lines and human induced pluripotent stem cells (iPSCs). Large-scale screening identified 499 lncRNA loci required for robust cellular growth, of which 89% showed growth-modifying function exclusively in one cell type. We further found that lncRNA knockdown can perturb complex transcriptional networks in a cell type–specific manner. These data underscore the functional importance and cell type specificity of many lncRNAs.

2010 ◽  
Vol 84 (12) ◽  
pp. 6139-6152 ◽  
Author(s):  
Amy L. Ellis ◽  
Zhenxun Wang ◽  
Xianming Yu ◽  
Janet E. Mertz

ABSTRACT We previously reported that the cellular protein ZEB1 can repress expression of the Epstein-Barr virus (EBV) BZLF1 gene in transient transfection assays by directly binding its promoter, Zp. We also reported that EBV containing a 2-bp substitution mutation in the ZEB-binding ZV element of Zp spontaneously reactivated out of latency into lytic replication at a higher frequency than did wild-type EBV. Here, using small interfering RNA (siRNA) and short hairpin RNA (shRNA) technologies, we definitively show that ZEB1 is, indeed, a key player in maintaining EBV latency in some epithelial and B-lymphocytic cell lines. However, in other EBV-positive epithelial and B-cell lines, another zinc finger E-box-binding protein, ZEB2/SIP1, is the key player. Both ZEB1 and ZEB2 can bind Zp via the ZV element. In EBV-positive cells containing only ZEB1, knockdown of ZEB1 led to viral reactivation out of latency, with synthesis of EBV immediate-early and early lytic gene products. However, in EBV-positive cells containing both ZEBs, ZEB2, not ZEB1, was the primary ZEB family member bound to Zp. Knockdown of ZEB2, but not ZEB1, led to EBV lytic reactivation. Thus, we conclude that either ZEB1 or ZEB2 can play a central role in the maintenance of EBV latency, doing so in a cell-type-dependent manner.


1994 ◽  
Vol 14 (1) ◽  
pp. 268-276 ◽  
Author(s):  
W Hsu ◽  
T K Kerppola ◽  
P L Chen ◽  
T Curran ◽  
S Chen-Kiang

NF-IL6 and AP-1 family transcription factors are coordinately induced by interleukin-6 (IL-6) in a cell-type-specific manner, suggesting that they mediate IL-6 signals in the nucleus. We show that the basic leucine zipper (bZIP) region of NF-IL6 mediates a direct association with the bZIP regions of Fos and Jun in vitro. This interaction does not depend on the presence of their cognate recognition DNA elements or the posttranslational modification of either partner. NF-IL6 homodimers can bind to both NF-IL6 and AP-1 sites, whereas Fos and Jun cannot bind to most NF-IL6 sites. Cross-family association with Fos or with Jun alters the DNA binding specificity of NF-IL6 and reduced its binding to NF-IL6 sites. NF-IL6 isoforms that differ in the site of translation initiation have distinct transcriptional activities. Activation of a reporter gene linked to the NF-IL6 site by NF-IL6 is repressed by Fos and by Jun in transient transfection assays. Thus, association with AP-1 results in repression of transcription activation by NF-IL6. The repression is NF-IL6 site dependent and may have a role in determining the promoter and cell type specificity in IL-6 signaling.


2020 ◽  
Author(s):  
Christina K. Kim ◽  
Mateo I. Sanchez ◽  
Paul Hoerbelt ◽  
Lief E. Fenno ◽  
Robert C. Malenka ◽  
...  

SUMMARYThe ability to record transient cellular events in the DNA or RNA of cells would enable precise, large-scale analysis, selection, and reprogramming of heterogeneous cell populations. Here we report a molecular technology for stable genetic tagging of cells that exhibit activity-related increases in intracellular calcium concentration (FLiCRE). We used FLiCRE to transcriptionally label activated neural ensembles in the nucleus accumbens of the mouse brain during brief stimulation of aversive inputs. Using single-cell RNA sequencing, we detected FLiCRE transcripts among the endogenous transcriptome, providing simultaneous readout of both cell-type and calcium activation history. We identified a cell-type in the nucleus accumbens activated downstream of long-range excitatory projections. Taking advantage of FLiCRE’s modular design, we expressed an optogenetic channel selectively in this cell-type, and showed that direct recruitment of this otherwise genetically-inaccessible population elicits behavioral aversion. The specificity and minute-resolution of FLiCRE enables molecularly-informed characterization, manipulation, and reprogramming of activated cellular ensembles.


2020 ◽  
Author(s):  
Brodie Fischbacher ◽  
Sarita Hedaya ◽  
Brigham J. Hartley ◽  
Zhongwei Wang ◽  
Gregory Lallos ◽  
...  

AbstractMonoclonalization refers to the isolation and expansion of a single cell derived from a cultured population. This is a valuable step in cell culture so as to minimize a cell line’s technical variability downstream of cell-altering events, such as reprogramming or gene editing, as well as for processes such as monoclonal antibody development. However, traditional methods for verifying clonality do not scale well, posing a critical obstacle to studies involving large cohorts. Without automated, standardized methods for assessing clonality post-hoc, methods involving monoclonalization cannot be reliably upscaled without exacerbating the technical variability of cell lines. We report the design of a deep learning workflow that automatically detects colony presence and identifies clonality from cellular imaging. The workflow, termed Monoqlo, integrates multiple convolutional neural networks and, critically, leverages the chronological directionality of the cell culturing process. Our algorithm design provides a fully scalable, highly interpretable framework, capable of analyzing industrial data volumes in under an hour using commodity hardware. In the present study, we focus on monoclonalization of human induced pluripotent stem cells (HiPSCs) as a case example. Monoqlo standardizes the monoclonalization process, enabling colony selection protocols to be infinitely upscaled while minimizing technical variability.


Cells ◽  
2022 ◽  
Vol 11 (1) ◽  
pp. 169
Author(s):  
Patrick D. Skelton ◽  
Valerie Tokars ◽  
Loukia Parisiadou

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease with a similar clinical presentation and progression to idiopathic Parkinson’s disease, and common variation is linked to disease risk. Recapitulation of the genotype in rodent models causes abnormal dopamine release and increases the susceptibility of dopaminergic neurons to insults, making LRRK2 a valuable model for understanding the pathobiology of Parkinson’s disease. It is also a promising druggable target with targeted therapies currently in development. LRRK2 mRNA and protein expression in the brain is highly variable across regions and cellular identities. A growing body of work has demonstrated that pathogenic LRRK2 mutations disrupt striatal synapses before the onset of overt neurodegeneration. Several substrates and interactors of LRRK2 have been identified to potentially mediate these pre-neurodegenerative changes in a cell-type-specific manner. This review discusses the effects of pathogenic LRRK2 mutations in striatal neurons, including cell-type-specific and pathway-specific alterations. It also highlights several LRRK2 effectors that could mediate the alterations to striatal function, including Rabs and protein kinase A. The lessons learned from improving our understanding of the pathogenic effects of LRRK2 mutations in striatal neurons will be applicable to both dissecting the cell-type specificity of LRRK2 function in the transcriptionally diverse subtypes of dopaminergic neurons and also increasing our understanding of basal ganglia development and biology. Finally, it will inform the development of therapeutics for Parkinson’s disease.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1862-1862
Author(s):  
David Spencer ◽  
Daniel R. George ◽  
Jeffery M. Klco ◽  
Timothy J. Ley

Abstract Somatic reprogramming captures the mutations present in individual cells and can yield induced pluripotent stem (iPS) cells that can be used to study these mutations in their native genetic context. iPS cells have been made using a variety of primary tissues and established cell lines, but to date there have been few examples of somatic reprogramming using primary cancer samples. Some studies have reported iPS cell generation using samples from patients with myeloproliferative neoplasms (Ye Z Blood 2009, Hosoi M Exp. Hematol. 2014), and another study successfully reprogrammed primary bone marrow cells from patients with myelodysplastic syndromes (MDS) (Kotini AG Nat. Biotech. 2015). However, it is not yet clear whether fully transformed human myeloid leukemia cells can be reprogrammed to an undifferentiated state. Here we describe the results of reprogramming experiments and subsequent genetic characterization of iPS clones produced from primary bone marrow and peripheral blood samples from adult human de novo AML patients. Our reprogramming approach involved in vitro culture of primary cells on Hs27 stroma with hematopoietic cytokines for 3-7 days, followed by transfer of 250,000 cells to stroma-free conditions for transduction with nonintegrating Sendai viruses expressing cMyc, OCT3/4, KLF4, and SOX2. Cells were then returned to AML culture conditions with stroma for 2-4 days before plating on mouse embryonic fibroblasts (MEF) in human embryonic stem (ES) cell media for 2-6 weeks. Individual clusters of cells with undifferentiated iPS cell colony morphology were then picked and expanded on either MEFs or feeder-free conditions. We performed 21 transductions using 8 peripheral blood and 13 bone marrow samples from 16 AML patients (i.e., multiple samples were attempted for some AMLs), which yielded 65 iPS clones from 9 of the 16 AML patients (56%) that were successfully expanded for genomic analysis. The remaining AMLs either produced no colonies (N=5), or clones that failed to expand after transferring from the original plate (N=2). Initial analysis of representative iPS clones (N=4) via flow cytometry demonstrated expression of the pluripotency markers SSEA-4 and TRA-1-60. Additional experiments to assess the pluripotency of these iPS lines are currently underway, including analysis of all clones via flow cytometry, RNA-sequencing, and teratoma formation assays. To determine the relationship between each iPS clone and the original AML samples used for reprogramming, we performed targeted sequencing for all somatic mutations identified from either whole-genome or exome sequencing. Analysis of each iPS clone for multiple patient-specific AML mutations (range 12-683) demonstrated that the reprogrammed cells were derived from 1 of 3 distinct cell types, depending on the sample. The most common type (N=1, 1, 3, 10, and 12 clones from 5 AMLs) possessed virtually no AML mutations (Figure 1A), suggesting that reprogramming occurred in a cell population that was unrelated to the tumor. Another 24 clones from 2 AML samples (N=1 and N=23) contained a subset of the AML-associated mutations (Figure 1B), but lacked common AML mutations that are generally cooperating 'hits', such as NPM1, and FLT3; for these samples, reprogramming probably occurred in a cell that was ancestral to the AML founding clone (i.e., a pre-leukemic cell). The final group of 14 clones from 2 AMLs (N=7 for both samples) contained the majority of AML-associated mutations in those samples, including canonical mutations in IDH1 and IDH2, and mutations in DNMT3A and RUNX1 (Figure 1C), implying that reprogramming occurred in the most prevalent AML subclone in the sample. Remarkably, for AML samples that yielded >1 iPS clone (N=6), all the iPS clones had the same set of mutations, suggesting that some of the cells in the sample were more "fit" for reprogramming than others. In conclusion, we have generated iPS cell lines from 9 primary AML samples, several of which contain canonical AML mutations. In this study, the majority of the reprogramming events took place in rare cells from clones that were not the most abundant cells in the sample. However, in one case, all iPS clones were derived from the most prevalent AML subclone in the sample. Future study of these iPS cell lines will provide insights into epigenetic dysregulation in cancer, and of the functional consequences of the mutational combinations that were "captured" via reprogramming. Disclosures No relevant conflicts of interest to declare.


Genes ◽  
2019 ◽  
Vol 10 (8) ◽  
pp. 593 ◽  
Author(s):  
Felipe Wendt Porto ◽  
Swapna Vidhur Daulatabad ◽  
Sarath Chandra Janga

Recent developments in our understanding of the interactions between long non-coding RNAs (lncRNAs) and cellular components have improved treatment approaches for various human diseases including cancer, vascular diseases, and neurological diseases. Although investigation of specific lncRNAs revealed their role in the metabolism of cellular RNA, our understanding of their contribution to post-transcriptional regulation is relatively limited. In this study, we explore the role of lncRNAs in modulating alternative splicing and their impact on downstream protein–RNA interaction networks. Analysis of alternative splicing events across 39 lncRNA knockdown and wildtype RNA-sequencing datasets from three human cell lines—HeLa (cervical cancer), K562 (myeloid leukemia), and U87 (glioblastoma)—resulted in the high-confidence (false discovery rate (fdr) < 0.01) identification of 11,630 skipped exon events and 5895 retained intron events, implicating 759 genes to be impacted at the post-transcriptional level due to the loss of lncRNAs. We observed that a majority of the alternatively spliced genes in a lncRNA knockdown were specific to the cell type. In tandem, the functions annotated to the genes affected by alternative splicing across each lncRNA knockdown also displayed cell-type specificity. To understand the mechanism behind this cell-type-specific alternative splicing pattern, we analyzed RNA-binding protein (RBP)–RNA interaction profiles across the spliced regions in order to observe cell-type-specific alternative splice event RBP binding preference. Despite limited RBP binding data across cell lines, alternatively spliced events detected in lncRNA perturbation experiments were associated with RBPs binding in proximal intron–exon junctions in a cell-type-specific manner. The cellular functions affected by alternative splicing were also affected in a cell-type-specific manner. Based on the RBP binding profiles in HeLa and K562 cells, we hypothesize that several lncRNAs are likely to exhibit a sponge effect in disease contexts, resulting in the functional disruption of RBPs and their downstream functions. We propose that such lncRNA sponges can extensively rewire post-transcriptional gene regulatory networks by altering the protein–RNA interaction landscape in a cell-type-specific manner.


2005 ◽  
Vol 86 (8) ◽  
pp. 2305-2314 ◽  
Author(s):  
Marian Wiegand ◽  
Sascha Bossow ◽  
Wolfgang J. Neubert

Induction of apoptosis during Sendai virus (SeV) infection has previously been documented to be triggered by initiator caspases (for strain F) or by a contribution of the cellular protein TIAR (T-cell-activated intracellular antigen-related) (for strain Z). Here, evidence was provided that both TIAR and caspases are simultaneously involved in apoptosis induction as a result of infection with SeV strain F. SeV F infection induced death in all tested cell lines, which could only be partially prevented through the pan-caspase inhibitor z-VAD-fmk. However, infection of seven different cell lines with the SeV mutant Fctr48z overexpressing a TIAR-sequestering RNA from the modified leader resulted in a cell type-dependent reduced cytopathic effect (CPE); in an earlier study a similar mutant derived from SeV Z was shown to prevent the induction of any CPE. Finally, blocking of caspases through z-VAD-fmk combined with Fctr48z infection led to complete abrogation of CPE, clearly demonstrating the existence of two separate mechanisms inducing cell death during SeV F infections. Interestingly, a cell type-specific interference between these two mechanisms could be detected during infection with the mutant virus Fctr48z: RNA transcribed from the mutated leader was able to trans-dominantly inhibit caspase-mediated apoptosis. Thus, virus-expressed factors enabling a well-balanced ratio of suppression and triggering of apoptosis seem to be essential for optimal virus replication.


2020 ◽  
Author(s):  
Kun Zhang ◽  
Yanbin Zhao

AbstractCell-based assays represent nearly half of all high-throughput screens currently conducted for risk assessment of environmental chemicals. However, the sensitivity and heterogeneity among cell lines has long been concerned but explored only in a limited manner. Here, we address this question by conducting a large scale transcriptomic analysis of the responses of discrete cell lines to specific small molecules. Our results illustrate heterogeneity of the extent and timing of responses among cell lines. Interestingly, high sensitivity and/or heterogeneity was found to be cell type-specific or universal depending on the different mechanism of actions of the compounds. Our data provide a novel insight into the understanding of cell-small molecule interactions and have substantial implications for the design, execution and interpretation of high-throughput screening assays.


2021 ◽  
Vol 118 (4) ◽  
pp. e2018861118
Author(s):  
Wentao Chen ◽  
Kurt Yun Mou ◽  
Paige Solomon ◽  
Rahul Aggarwal ◽  
Kevin K. Leung ◽  
...  

MYC is a powerful transcription factor overexpressed in many human cancers including B cell and prostate cancers. Antibody therapeutics are exciting opportunities to attack cancers but require knowledge of surface proteins that change due to oncogene expression. To identify how MYC overexpression remodels the cell surface proteome in a cell autologous fashion and in different cell types, we investigated the impact of MYC overexpression on 800 surface proteins in three isogenic model cell lines either of B cell or prostate cell origin engineered to have high or low MYC levels. We found that MYC overexpression resulted in dramatic remodeling (both up- and down-regulation) of the cell surfaceome in a cell type-dependent fashion. We found systematic and large increases in distinct sets of >80 transporters including nucleoside transporters and nutrient transporters making cells more sensitive to toxic nucleoside analogs like cytarabine, commonly used for treating hematological cancers. Paradoxically, MYC overexpression also increased expression of surface proteins driving cell turnover such as TNFRSF10B, also known as death receptor 5, and immune cell attacking signals such as the natural killer cell activating ligand NCR3LG1, also known as B7-H6. We generated recombinant antibodies to these two targets and verified their up-regulation in MYC overexpression cell lines and showed they were sensitive to bispecific T cell engagers (BiTEs). Our studies demonstrate how MYC overexpression leads to dramatic bidirectional remodeling of the surfaceome in a cell type-dependent but functionally convergent fashion and identify surface targets or combinations thereof as possible candidates for cytotoxic metabolite or immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document