Matching Items (14)
Description

Linnorm is a novel normalization and transformation method for the analysis of single cell RNA sequencing (scRNA-seq) data. Linnorm is developed to remove technical noises and simultaneously preserve biological variations in scRNA-seq data, such that existing statistical methods can be improved. Using real scRNA-seq data, we compared Linnorm with existing

Linnorm is a novel normalization and transformation method for the analysis of single cell RNA sequencing (scRNA-seq) data. Linnorm is developed to remove technical noises and simultaneously preserve biological variations in scRNA-seq data, such that existing statistical methods can be improved. Using real scRNA-seq data, we compared Linnorm with existing normalization methods, including NODES, SAMstrt, SCnorm, scran, DESeq and TMM. Linnorm shows advantages in speed, technical noise removal and preservation of cell heterogeneity, which can improve existing methods in the discovery of novel subtypes, pseudo-temporal ordering of cells, clustering analysis, etc. Linnorm also performs better than existing DEG analysis methods, including BASiCS, NODES, SAMstrt, Seurat and DESeq2, in false positive rate control and accuracy.

ContributorsYip, Shun H. (Author) / Wang, Panwen (Author) / Kocher, Jean-Pierre A. (Author) / Sham, Pak Chung (Author) / Wang, Junwen (Author) / College of Health Solutions (Contributor)
Created2017-09-18
128638-Thumbnail Image.png
Description

It remains challenging to predict regulatory variants in particular tissues or cell types due to highly context-specific gene regulation. By connecting large-scale epigenomic profiles to expression quantitative trait loci (eQTLs) in a wide range of human tissues/cell types, we identify critical chromatin features that predict variant regulatory potential. We present

It remains challenging to predict regulatory variants in particular tissues or cell types due to highly context-specific gene regulation. By connecting large-scale epigenomic profiles to expression quantitative trait loci (eQTLs) in a wide range of human tissues/cell types, we identify critical chromatin features that predict variant regulatory potential. We present cepip, a joint likelihood framework, for estimating a variant’s regulatory probability in a context-dependent manner. Our method exhibits significant GWAS signal enrichment and is superior to existing cell type-specific methods. Furthermore, using phenotypically relevant epigenomes to weight the GWAS single-nucleotide polymorphisms, we improve the statistical power of the gene-based association test.

ContributorsLi, Mulin Jun (Author) / Li, Miaoxin (Author) / Liu, Zipeng (Author) / Yan, Bin (Author) / Pan, Zhicheng (Author) / Huang, Dandan (Author) / Liang, Qian (Author) / Ying, Dingge (Author) / Xu, Feng (Author) / Yao, Hongcheng (Author) / Wang, Panwen (Author) / Kocher, Jean-Pierre A. (Author) / Xia, Zhengyuan (Author) / Sham, Pak Chung (Author) / Liu, Jun S. (Author) / Wang, Junwen (Author) / College of Health Solutions (Contributor)
Created2017-03-16
128573-Thumbnail Image.png
Description

Two distinct monocyte (Mo)/macrophage (Mp) subsets (Ly6Clow and Ly6Chi) orchestrate cardiac recovery process following myocardial infarction (MI). Prostaglandin (PG) E2 is involved in the Mo/Mp-mediated inflammatory response, however, the role of its receptors in Mos/Mps in cardiac healing remains to be determined. Here we show that pharmacological inhibition or gene

Two distinct monocyte (Mo)/macrophage (Mp) subsets (Ly6Clow and Ly6Chi) orchestrate cardiac recovery process following myocardial infarction (MI). Prostaglandin (PG) E2 is involved in the Mo/Mp-mediated inflammatory response, however, the role of its receptors in Mos/Mps in cardiac healing remains to be determined. Here we show that pharmacological inhibition or gene ablation of the Ep3 receptor in mice suppresses accumulation of Ly6Clow Mos/Mps in infarcted hearts. Ep3 deletion in Mos/Mps markedly attenuates healing after MI by reducing neovascularization in peri-infarct zones. Ep3 deficiency diminishes CX3C chemokine receptor 1 (CX3CR1) expression and vascular endothelial growth factor (VEGF) secretion in Mos/Mps by suppressing TGFβ1 signaling and subsequently inhibits Ly6Clow Mos/Mps migration and angiogenesis. Targeted overexpression of Ep3 receptors in Mos/Mps improves wound healing by enhancing angiogenesis. Thus, the PGE2/Ep3 axis promotes cardiac healing after MI by activating reparative Ly6Clow Mos/Mps, indicating that Ep3 receptor activation may be a promising therapeutic target for acute MI.

ContributorsTang, Juan (Author) / Shen, Yujun (Author) / Chen, Guilin (Author) / Wan, Qiangyou (Author) / Wang, Kai (Author) / Zhang, Jian (Author) / Qin, Jing (Author) / Liu, Guizhu (Author) / Zuo, Shengkai (Author) / Tao, Bo (Author) / Yu, Yu (Author) / Wang, Junwen (Author) / Lazarus, Michael (Author) / Yu, Ying (Author) / College of Health Solutions (Contributor)
Created2017-03-03
128532-Thumbnail Image.png
Description

Accumulating data from genome-wide association studies (GWAS) have provided a collection of novel candidate genes associated with complex diseases, such as atherosclerosis. We identified an atherosclerosis-associated single-nucleotide polymorphism (SNP) located in the intron of the long noncoding RNA (lncRNA) LINC00305 by searching the GWAS database. Although the function of LINC00305

Accumulating data from genome-wide association studies (GWAS) have provided a collection of novel candidate genes associated with complex diseases, such as atherosclerosis. We identified an atherosclerosis-associated single-nucleotide polymorphism (SNP) located in the intron of the long noncoding RNA (lncRNA) LINC00305 by searching the GWAS database. Although the function of LINC00305 is unknown, we found that LINC00305 expression is enriched in atherosclerotic plaques and monocytes. Overexpression of LINC00305 promoted the expression of inflammation-associated genes in THP-1 cells and reduced the expression of contractile markers in co-cultured human aortic smooth muscle cells (HASMCs). We showed that overexpression of LINC00305 activated nuclear factor-kappa beta (NF-κB) and that inhibition of NF-κB abolished LINC00305-mediated activation of cytokine expression. Mechanistically, LINC00305 interacted with lipocalin-1 interacting membrane receptor (LIMR), enhanced the interaction of LIMR and aryl-hydrocarbon receptor repressor (AHRR), and promoted protein expression as well as nuclear localization of AHRR. Moreover, LINC00305 activated NF-κB exclusively in the presence of LIMR and AHRR. In light of these findings, we propose that LINC00305 promotes monocyte inflammation by facilitating LIMR and AHRR cooperation and the AHRR activation, which eventually activates NF-κB, thereby inducing HASMC phenotype switching.

ContributorsZhang, Dan-Dan (Author) / Wang, Wen-Tian (Author) / Xiong, Jian (Author) / Xie, Xue-Min (Author) / Cui, Shen-Shen (Author) / Zhao, Zhi-Guo (Author) / Li, Mulin Jun (Author) / Zhang, Zhu-Qin (Author) / Hao, De-Long (Author) / Zhao, Xiang (Author) / Li, Yong-Jun (Author) / Wang, Junwen (Author) / Chen, Hou-Zao (Author) / Lv, Xiang (Author) / Liu, De-Pei (Author) / College of Health Solutions (Contributor)
Created2017-04-10