Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Chun Ye is active.

Publication


Featured researches published by Chun Ye.


Nature Biotechnology | 2005

Assessing computational tools for the discovery of transcription factor binding sites

Martin Tompa; Nan Li; Timothy L. Bailey; George M. Church; Bart De Moor; Eleazar Eskin; Alexander V. Favorov; Martin C. Frith; Yutao Fu; W. James Kent; Vsevolod J. Makeev; Andrei A. Mironov; William Stafford Noble; Giulio Pavesi; Mireille Régnier; Nicolas Simonis; Saurabh Sinha; Gert Thijs; Jacques van Helden; Mathias Vandenbogaert; Zhiping Weng; Christopher T. Workman; Chun Ye; Zhou Zhu

The prediction of regulatory elements is a problem where computational methods offer great hope. Over the past few years, numerous tools have become available for this task. The purpose of the current assessment is twofold: to provide some guidance to users regarding the accuracy of currently available tools in various settings, and to provide a benchmark of data sets for assessing future tools.


Science | 2014

Polarization of the Effects of Autoimmune and Neurodegenerative Risk Alleles in Leukocytes

Towfique Raj; Katie Rothamel; Chun Ye; Mark Lee; Joseph M. Replogle; Ting Feng; Michelle Lee; Natasha Asinovski; Irene Y. Frohlich; Selina Imboywa; Alina Von Korff; Yukinori Okada; Nikolaos A. Patsopoulos; Scott Davis; Cristin McCabe; Hyun Il Paik; Gyan Srivastava; Soumya Raychaudhuri; David A. Hafler; Daphne Koller; Aviv Regev; Nir Hacohen; Diane Mathis; Christophe Benoist; Barbara E. Stranger; Philip L. De Jager

Immunogenetic Variation Many genetic variants have been implicated in disease but their effects in function across tissues and cell-types remain to be resolved. Raj et al. (p. 519) present an analysis of expression quantative trait loci (eQTL) measuring messenger RNA levels and examined correlations between genotypes and gene expression in purified monocytes and T cells in healthy individuals of European, African, and Asian descent. Most, but not all, of the eQTLs and their effects on expression were shared between the populations, as well as a substantial proportion between the cell types. Links were found with disease-associated variants and loci that previous genome-wide analyses have implicated in neurodegenerative and autoimmune diseases. Genetic variants affecting gene expression show both shared and specific immune cell type effects. To extend our understanding of the genetic basis of human immune function and dysfunction, we performed an expression quantitative trait locus (eQTL) study of purified CD4+ T cells and monocytes, representing adaptive and innate immunity, in a multi-ethnic cohort of 461 healthy individuals. Context-specific cis- and trans-eQTLs were identified, and cross-population mapping allowed, in some cases, putative functional assignment of candidate causal regulatory variants for disease-associated loci. We note an over-representation of T cell–specific eQTLs among susceptibility alleles for autoimmune diseases and of monocyte-specific eQTLs among Alzheimer’s and Parkinson’s disease variants. This polarization implicates specific immune cell types in these diseases and points to the need to identify the cell-autonomous effects of disease susceptibility variants.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Generation of knock-in primary human T cells using Cas9 ribonucleoproteins

Kathrin Schumann; Steven Lin; Eric Boyer; Dimitre R. Simeonov; Meena Subramaniam; Rachel E. Gate; Genevieve E. Haliburton; Chun Ye; Jeffrey A. Bluestone; Jennifer A. Doudna; Alexander Marson

Significance T-cell genome engineering holds great promise for cancer immunotherapies and cell-based therapies for HIV, primary immune deficiencies, and autoimmune diseases, but genetic manipulation of human T cells has been inefficient. We achieved efficient genome editing by delivering Cas9 protein pre-assembled with guide RNAs. These active Cas9 ribonucleoproteins (RNPs) enabled successful Cas9-mediated homology-directed repair in primary human T cells. Cas9 RNPs provide a programmable tool to replace specific nucleotide sequences in the genome of mature immune cells—a longstanding goal in the field. These studies establish Cas9 RNP technology for diverse experimental and therapeutic genome engineering applications in primary human T cells. T-cell genome engineering holds great promise for cell-based therapies for cancer, HIV, primary immune deficiencies, and autoimmune diseases, but genetic manipulation of human T cells has been challenging. Improved tools are needed to efficiently “knock out” genes and “knock in” targeted genome modifications to modulate T-cell function and correct disease-associated mutations. CRISPR/Cas9 technology is facilitating genome engineering in many cell types, but in human T cells its efficiency has been limited and it has not yet proven useful for targeted nucleotide replacements. Here we report efficient genome engineering in human CD4+ T cells using Cas9:single-guide RNA ribonucleoproteins (Cas9 RNPs). Cas9 RNPs allowed ablation of CXCR4, a coreceptor for HIV entry. Cas9 RNP electroporation caused up to ∼40% of cells to lose high-level cell-surface expression of CXCR4, and edited cells could be enriched by sorting based on low CXCR4 expression. Importantly, Cas9 RNPs paired with homology-directed repair template oligonucleotides generated a high frequency of targeted genome modifications in primary T cells. Targeted nucleotide replacement was achieved in CXCR4 and PD-1 (PDCD1), a regulator of T-cell exhaustion that is a validated target for tumor immunotherapy. Deep sequencing of a target site confirmed that Cas9 RNPs generated knock-in genome modifications with up to ∼20% efficiency, which accounted for up to approximately one-third of total editing events. These results establish Cas9 RNP technology for diverse experimental and therapeutic genome engineering applications in primary human T cells.


Science | 2014

Common genetic variants modulate pathogen-sensing responses in human dendritic cells.

Mark Lee; Chun Ye; Alexandra-Chloé Villani; Towfique Raj; Weibo Li; Thomas Eisenhaure; Selina Imboywa; Portia Chipendo; F. Ann Ran; Kamil Slowikowski; Lucas D. Ward; Cristin McCabe; Michelle Lee; Irene Y. Frohlich; David A. Hafler; Manolis Kellis; Soumya Raychaudhuri; Feng Zhang; Barbara E. Stranger; Christophe Benoist; Philip L. De Jager; Aviv Regev; Nir Hacohen

Introduction Variation in an individual’s response to environmental factors is likely to influence susceptibility to complex human diseases. The genetic basis of such variation is poorly understood. Here, we identify natural genetic variants that underlie variation in the host innate immune response to infection and analyze the mechanisms by which such variants alter these responses. Identifying the genetic basis of variability in the host response to pathogens. A cohort of 534 individuals donated blood for (a) genotyping of common DNA variants and (b) isolation of immune DCs. DCs were stimulated with viral and bacterial components, and the variability in individuals’ gene expression responses was mapped to specific DNA variants, which were then shown to affect binding of particular transcription factors. Methods We derived dendritic cells (DCs) from peripheral blood monocytes of healthy individuals (295 Caucasians, 122 African Americans, 117 East Asians) and stimulated them with Escherichia coli lipopolysaccharide (LPS), influenza virus, or the cytokine interferon-β (IFN-β) to generate 1598 transcriptional profiles. We genotyped each of these individuals at sites of common genetic variation and identified the genetic variants that best explain variation in gene expression and gene induction between individuals. We then tested mechanistic predictions from these associations using synthetic promoter constructs and genome engineering. Results We identified 264 loci containing genetic variants associated with variation in absolute gene expression in human DCs, of which 121 loci were associated with variation in the induction of gene expression by one or more stimuli. Fine-mapping identified candidate causal single-nucleotide polymorphisms (SNPs) associated with expression variance, and deeper functional experiments localized three of these SNPs to the binding sites of stimulus-activated transcription factors. We also identified a cis variant in the transcription factor, IRF7, associated in trans with the induction of a module of antiviral genes in response to influenza infection. Of the identified genetic variants, 35 were also associated with autoimmune or infectious disease loci found by genome-wide association studies. Discussion The genetic variants we uncover and the molecular basis for their action provide mechanistic explanations and principles for how the innate immune response to pathogens and cytokines varies across individuals. Our results also link disease-associated variants to specific immune pathways in DCs, which provides greater insight into mechanisms underlying complex human phenotypes. Extending our approach to many immune cell types and pathways will provide a global map linking human genetic variants to specific immunological processes. Immune Variation It is difficult to determine the mechanistic consequences of context-dependent genetic variants, some of which may be related to disease (see the Perspective by Gregersen). Two studies now report on the effects of stimulating immunological monocytes and dendritic cells with proteins that can elicit a response to bacterial or viral infection and assess the functional links between genetic variants and profiles of gene expression. M. N. Lee et al. (10.1126/science.1246980) analyzed the expression of more than 400 genes, in dendritic cells from 534 healthy subjects, which revealed how expression quantitative trait loci (eQTLs) affect gene expression within the interferon-β and the Toll-like receptor 3 and 4 pathways. Fairfax et al. (10.1126/science.1246949) performed a genome-wide analysis to show that many eQTLs affected monocyte gene expression in a stimulus- or time-specific manner. Mapping of human host-pathogen gene-by-environment interactions identifies pathogen-specific loci. [Also see Perspective by Gregersen] Little is known about how human genetic variation affects the responses to environmental stimuli in the context of complex diseases. Experimental and computational approaches were applied to determine the effects of genetic variation on the induction of pathogen-responsive genes in human dendritic cells. We identified 121 common genetic variants associated in cis with variation in expression responses to Escherichia coli lipopolysaccharide, influenza, or interferon-β (IFN-β). We localized and validated causal variants to binding sites of pathogen-activated STAT (signal transducer and activator of transcription) and IRF (IFN-regulatory factor) transcription factors. We also identified a common variant in IRF7 that is associated in trans with type I IFN induction in response to influenza infection. Our results reveal common alleles that explain interindividual variation in pathogen sensing and provide functional annotation for genetic variants that alter susceptibility to inflammatory diseases.


Nature Genetics | 2013

Mutations causing medullary cystic kidney disease type 1 lie in a large VNTR in MUC1 missed by massively parallel sequencing

Andrew Kirby; Andreas Gnirke; David B. Jaffe; Veronika Barešová; Nathalie Pochet; Brendan Blumenstiel; Chun Ye; Daniel Aird; Christine Stevens; James Robinson; Moran N. Cabili; Irit Gat-Viks; Edward Kelliher; Riza Daza; Matthew DeFelice; Helena Hůlková; Jana Sovová; Petr Vylet’al; Corinne Antignac; Mitchell Guttman; Robert E. Handsaker; Danielle Perrin; Scott Steelman; Snaevar Sigurdsson; Steven J. Scheinman; Carrie Sougnez; Kristian Cibulskis; Melissa Parkin; Todd Green; Elizabeth Rossin

Although genetic lesions responsible for some mendelian disorders can be rapidly discovered through massively parallel sequencing of whole genomes or exomes, not all diseases readily yield to such efforts. We describe the illustrative case of the simple mendelian disorder medullary cystic kidney disease type 1 (MCKD1), mapped more than a decade ago to a 2-Mb region on chromosome 1. Ultimately, only by cloning, capillary sequencing and de novo assembly did we find that each of six families with MCKD1 harbors an equivalent but apparently independently arising mutation in sequence markedly under-represented in massively parallel sequencing data: the insertion of a single cytosine in one copy (but a different copy in each family) of the repeat unit comprising the extremely long (∼1.5–5 kb), GC-rich (>80%) coding variable-number tandem repeat (VNTR) sequence in the MUC1 gene encoding mucin 1. These results provide a cautionary tale about the challenges in identifying the genes responsible for mendelian, let alone more complex, disorders through massively parallel sequencing.


Genetics | 2008

Accurate discovery of expression quantitative trait loci under confounding from spurious and genuine regulatory hotspots.

Hyun Min Kang; Chun Ye; Eleazar Eskin

In genomewide mapping of expression quantitative trait loci (eQTL), it is widely believed that thousands of genes are trans-regulated by a small number of genomic regions called “regulatory hotspots,” resulting in “trans-regulatory bands” in an eQTL map. As several recent studies have demonstrated, technical confounding factors such as batch effects can complicate eQTL analysis by causing many spurious associations including spurious regulatory hotspots. Yet little is understood about how these technical confounding factors affect eQTL analyses and how to correct for these factors. Our analysis of data sets with biological replicates suggests that it is this intersample correlation structure inherent in expression data that leads to spurious associations between genetic loci and a large number of transcripts inducing spurious regulatory hotspots. We propose a statistical method that corrects for the spurious associations caused by complex intersample correlation of expression measurements in eQTL mapping. Applying our intersample correlation emended (ICE) eQTL mapping method to mouse, yeast, and human identifies many more cis associations while eliminating most of the spurious trans associations. The concordances of cis and trans associations have consistently increased between different replicates, tissues, and populations, demonstrating the higher accuracy of our method to identify real genetic effects.


Nature Immunology | 2016

A tissue checkpoint regulates type 2 immunity

Steven J. Van Dyken; Jesse C. Nussbaum; Jinwoo Lee; Ari B. Molofsky; Hong-Erh Liang; Joshua L. Pollack; Rachel E. Gate; Genevieve E. Haliburton; Chun Ye; Alexander Marson; David J. Erle; Richard M. Locksley

Group 2 innate lymphoid cells (ILC2s) and CD4+ type 2 helper T cells (TH2 cells) are defined by their similar effector cytokines, which together mediate the features of allergic immunity. We found that tissue ILC2s and TH2 cells differentiated independently but shared overlapping effector function programs that were mediated by exposure to the tissue-derived cytokines interleukin 25 (IL-25), IL-33 and thymic stromal lymphopoietin (TSLP). Loss of these three tissue signals did not affect lymph node priming, but abrogated the terminal differentiation of effector TH2 cells and adaptive lung inflammation in a T cell–intrinsic manner. Our findings suggest a mechanism by which diverse perturbations can activate type 2 immunity and reveal a shared local-tissue-elicited checkpoint that can be exploited to control both innate and adaptive allergic inflammation.


Scientific Reports | 2017

CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells.

Levi J. Rupp; Kathrin Schumann; Kole T. Roybal; Rachel E. Gate; Chun Ye; Wendell A. Lim; Alexander Marson

Immunotherapies with chimeric antigen receptor (CAR) T cells and checkpoint inhibitors (including antibodies that antagonize programmed cell death protein 1 [PD-1]) have both opened new avenues for cancer treatment, but the clinical potential of combined disruption of inhibitory checkpoints and CAR T cell therapy remains incompletely explored. Here we show that programmed death ligand 1 (PD-L1) expression on tumor cells can render human CAR T cells (anti-CD19 4-1BBζ) hypo-functional, resulting in impaired tumor clearance in a sub-cutaneous xenograft model. To overcome this suppressed anti-tumor response, we developed a protocol for combined Cas9 ribonucleoprotein (Cas9 RNP)-mediated gene editing and lentiviral transduction to generate PD-1 deficient anti-CD19 CAR T cells. Pdcd1 (PD-1) disruption augmented CAR T cell mediated killing of tumor cells in vitro and enhanced clearance of PD-L1+ tumor xenografts in vivo. This study demonstrates improved therapeutic efficacy of Cas9-edited CAR T cells and highlights the potential of precision genome engineering to enhance next-generation cell therapies.


PLOS Genetics | 2013

Effectively Identifying eQTLs from Multiple Tissues by Combining Mixed Model and Meta-analytic Approaches

Jae Hoon Sul; Buhm Han; Chun Ye; Ted Choi; Eleazar Eskin

Gene expression data, in conjunction with information on genetic variants, have enabled studies to identify expression quantitative trait loci (eQTLs) or polymorphic locations in the genome that are associated with expression levels. Moreover, recent technological developments and cost decreases have further enabled studies to collect expression data in multiple tissues. One advantage of multiple tissue datasets is that studies can combine results from different tissues to identify eQTLs more accurately than examining each tissue separately. The idea of aggregating results of multiple tissues is closely related to the idea of meta-analysis which aggregates results of multiple genome-wide association studies to improve the power to detect associations. In principle, meta-analysis methods can be used to combine results from multiple tissues. However, eQTLs may have effects in only a single tissue, in all tissues, or in a subset of tissues with possibly different effect sizes. This heterogeneity in terms of effects across multiple tissues presents a key challenge to detect eQTLs. In this paper, we develop a framework that leverages two popular meta-analysis methods that address effect size heterogeneity to detect eQTLs across multiple tissues. We show by using simulations and multiple tissue data from mouse that our approach detects many eQTLs undetected by traditional eQTL methods. Additionally, our method provides an interpretation framework that accurately predicts whether an eQTL has an effect in a particular tissue.


PLOS Computational Biology | 2009

Using Network Component Analysis to Dissect Regulatory Networks Mediated by Transcription Factors in Yeast

Chun Ye; Simon J. Galbraith; James C. Liao; Eleazar Eskin

Understanding the relationship between genetic variation and gene expression is a central question in genetics. With the availability of data from high-throughput technologies such as ChIP-Chip, expression, and genotyping arrays, we can begin to not only identify associations but to understand how genetic variations perturb the underlying transcription regulatory networks to induce differential gene expression. In this study, we describe a simple model of transcription regulation where the expression of a gene is completely characterized by two properties: the concentrations and promoter affinities of active transcription factors. We devise a method that extends Network Component Analysis (NCA) to determine how genetic variations in the form of single nucleotide polymorphisms (SNPs) perturb these two properties. Applying our method to a segregating population of Saccharomyces cerevisiae, we found statistically significant examples of trans-acting SNPs located in regulatory hotspots that perturb transcription factor concentrations and affinities for target promoters to cause global differential expression and cis-acting genetic variations that perturb the promoter affinities of transcription factors on a single gene to cause local differential expression. Although many genetic variations linked to gene expressions have been identified, it is not clear how they perturb the underlying regulatory networks that govern gene expression. Our work begins to fill this void by showing that many genetic variations affect the concentrations of active transcription factors in a cell and their affinities for target promoters. Understanding the effects of these perturbations can help us to paint a more complete picture of the complex landscape of transcription regulation. The software package implementing the algorithms discussed in this work is available as a MATLAB package upon request.

Collaboration


Dive into the Chun Ye's collaboration.

Top Co-Authors

Avatar

Eleazar Eskin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel E. Gate

University of California

View shared research outputs
Top Co-Authors

Avatar

Aviv Regev

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Boyer

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge