scholarly journals Coexpression enrichment analysis at the single-cell level reveals convergent defects in neural progenitor cells and their cell-type transitions in neurodevelopmental disorders

2020 ◽  
Vol 30 (6) ◽  
pp. 835-848 ◽  
Author(s):  
Kaifang Pang ◽  
Li Wang ◽  
Wei Wang ◽  
Jian Zhou ◽  
Chao Cheng ◽  
...  
Author(s):  
Zilong Zhang ◽  
Feifei Cui ◽  
Chen Lin ◽  
Lingling Zhao ◽  
Chunyu Wang ◽  
...  

Abstract Single-cell RNA sequencing (scRNA-seq) has enabled us to study biological questions at the single-cell level. Currently, many analysis tools are available to better utilize these relatively noisy data. In this review, we summarize the most widely used methods for critical downstream analysis steps (i.e. clustering, trajectory inference, cell-type annotation and integrating datasets). The advantages and limitations are comprehensively discussed, and we provide suggestions for choosing proper methods in different situations. We hope this paper will be useful for scRNA-seq data analysts and bioinformatics tool developers.


2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Ji heon Rhim ◽  
Xiangjian Luo ◽  
Dongbing Gao ◽  
Xiaoyun Xu ◽  
Tieling Zhou ◽  
...  

2018 ◽  
Author(s):  
Hengxing Ba ◽  
Datao Wang ◽  
Weiyao Wu ◽  
Hongmei Sun ◽  
Chunyi Li

AbstractAntler regeneration, a stem cell-based epimorphic process, has potential as a valuable model for regenerative medicine. A pool of antler stem cells (ASCs) for antler development is located in the antlerogenic periosteum (AP). However, whether this ASC pool is homogenous or heterogeneous has not been fully evaluated. In this study, we produced a comprehensive transcriptome dataset at the single-cell level for the ASCs based on the 10x Genomics platform (scRNA-seq). A total of 4,565 ASCs were sequenced and classified into a large cell cluster, indicating that the ASCs resident in the AP are likely to be a homogeneous population. The scRNA-seq data revealed that tumor-related genes were highly expressed in these homogeneous ASCs: i.e. TIMP1, TMSB10, LGALS1, FTH1, VIM, LOC110126017 and S100A4. Results of screening for stem cell markers suggest that the ASCs may be considered as a special type of stem cell between embryonic (CD9) and adult (CD29, CD90, NPM1 and VIM) stem cells. Our results provide the first comprehensive transcriptome analysis at the single-cell level for the ASCs, and identified only one major cell type resident in the AP and some key stem cell genes, which may hold the key to why antlers, the unique mammalian organ, can fully regenerate once lost.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5120-5120
Author(s):  
Sophia Miliara ◽  
Bogumil Kaczkowski ◽  
Takahiro Suzuki ◽  
Huthayfa Mujahed ◽  
Maasaki Furuno ◽  
...  

Abstract Acute Myeloid Leukemia (AML) is the most common myeloid leukemia in adults. Although substantial progress has been made in recent years, the long-term prognosis for patients remains poor which is mainly due to the dated treatments that consist of cytotoxic drugs with low specificity. AML is a clonal disease with multiple co-existing clones in each patient. Often, patients that initially respond to treatment may develop resistance due to lingering leukemic stem cells (LSC), or sub-clones that survive the treatment and cause a relapse. Therefore, novel therapeutic strategies are needed to fully eradicate all leukemic cells. AML has a strong epigenetic component meaning mutations in genes encoding epigenetic regulators are frequently acquired during early AML development, and are present in the initiating clones. Thus, targeting the epigenetic machinery may offer a new avenue for AML treatment. Among the newer epigenetic drugs are BET inhibitors, which bind reversibly to bromodomains of BRD proteins and prevent protein-protein interactions with acetylated histones and transcriptions factors. One of the most promising BET inhibitors is OTX015, which has already been in Phase II clinical trials for AML in the U.S. (Braun & Gardin, Expert Opinion on Investigational Drugs, 2017). We aim to analyze the heterogeneous response to OTX015 in AML, and normal stem/progenitor, cells in order to dissect the BET-inhibitor response. The main focus is the specific transcriptional signatures at promoters and enhancers as enhancers, and especially super-enhancers, have previously been shown to be sensitive to BET-inhibitors (Loven et al, Cell, 2013). To this effect, we have established a protocol that allowed for the transcriptional profiling of single cells from AML patients that were at different differentiation stages, using FACS- sorting. The patients were obtained from the Swedish Acute Leukemia Registry. To decrease population heterogeneity, the project focused on distinct subgroups of AML that previously has been shown to be sensitive for BET inhibitors. The different isolated AML, and normal progenitor populations, were exposed to OTX015 for 48hrs, and processed with both bulk transcriptional profiling of the general cell population response, and single cell profiling to analyze cell heterogeneity, and single cell response. For the transcriptional profiling, we utilized a unique technique called Cap Analysis of Gene Expression (CAGE), a powerful 5' start profiling technology, that allows for the identification of the transcription start site at base pair resolution, and determination of enhancer activity based on enhancer RNA expression. The single cell profiling was performed using C1 CAGE, which is a single-cell implementation CAGE (Kouno et al, bioRxiv 330845, 2018).We envision that the heterogenic transcriptional drug response, on the single-cell level, in AML and normal stem/progenitor cells will lead to the identification of key genes and pathways involved in the differential drug response. Additionally, the application of CAGE technology will lead to discovery of specific transcriptional signatures at promoters and enhancers that may be predictive of drug resistance. Clinical significance: Leukemic cell heterogeneity remains the main problem in AML, as chemotherapy often fails to completely eradicate all AML sub-clones including LSC, leading to relapses and high mortality of the disease. This study will shed light to the unique features of AML cell heterogeneity and how their drug response differs, not only between AML cells, but also between AML cells and their normal counterparts, on the single-cell level, based on the response to OTX015. The significance will be two-fold: the in-depth characterization of the features in AML populations and normal cells, and the potential this study will provide for novel, more targeted, combination treatments in AML. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 24 ◽  
pp. 129-133 ◽  
Author(s):  
Juanjiangmeng Du ◽  
Sean Simmons ◽  
Andreas Brunklaus ◽  
Xian Adiconis ◽  
Cynthia C. Hession ◽  
...  

2020 ◽  
Author(s):  
Kaifang Pang ◽  
Li Wang ◽  
Wei Wang ◽  
Jian Zhou ◽  
Chao Cheng ◽  
...  

AbstractRecent large-scale sequencing studies have identified a great number of genes whose disruptions cause neurodevelopmental disorders (NDDs). However, cell-type-specific functions of NDD genes and their contributions to NDD pathology are unclear. Here, we integrated NDD genetics with single-cell RNA sequencing data to identify cell-type and temporal convergence of genes involved in different NDDs. By assessing the co-expression enrichment pattern of various NDD gene sets, we identified mid-fetal cortical neural progenitor cell development—more specifically, ventricular radial glia-to-intermediate progenitor cell transition at gestational week 10—as a key convergent point in autism spectrum disorder (ASD) and epilepsy. Integrated gene ontology-based analyses further revealed that ASD genes function as upstream regulators to activate neural differentiation and inhibit cell cycle during the transition, whereas epilepsy genes function as downstream effectors in the same processes, offering a potential explanation for the high comorbidity rate of the two disorders. Together, our study provides a framework for investigating the cell-type-specific pathophysiology of NDDs.


Blood ◽  
1997 ◽  
Vol 90 (6) ◽  
pp. 2282-2292 ◽  
Author(s):  
Ole J. Borge ◽  
Veslemøy Ramsfjell ◽  
Li Cui ◽  
Sten E.W. Jacobsen

Purified primitive progenitor/stem cells from bone marrow represent likely target populations for ex vivo expansion of stem cells to be used in high-dose chemotherapy or gene therapy. Whereas such primitive progenitor cells require combined stimulation by multiple cytokines for growth, some cytokines selectively promote viability rather than growth when acting individually. We investigated here for the first time the direct effects of cytokines on survival of primitive CD34+CD38− human bone marrow progenitor cells at the single-cell level. Interleukin-3 (IL-3) and the ligands for c-kit (KL) and flt3 (FL) had direct and selective viability-promoting effects on a small fraction of CD34+CD38− but not CD34+CD38+ progenitor cells. Interestingly, the recently cloned thrombopoietin (Tpo), although stimulating little growth, kept most CD34+CD38− progenitors viable after prolonged culture, maintaining twofold and fourfold more progenitors viable than KL and IL-3, respectively. A high fraction of these progenitors had a combined myeloid and erythroid differentiation potential, as well as capacity for prolonged production of progenitor cells under stroma-independent conditions. In addition, Tpo promoted viability of CD34+CD38− long-term culture-initiating cells, further supporting the idea that Tpo promotes viability of primitive human progenitor cells. Finally, Tpo suppressed apoptosis of CD34+CD38− cells in culture. Thus, the present studies show a novel effect of Tpo, implicating a potential role of this cytokine in maintaining quiescent primitive human progenitor cells viable.


Sign in / Sign up

Export Citation Format

Share Document