Network


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

Hotspot


Dive into the research topics where Chongyuan Luo is active.

Publication


Featured researches published by Chongyuan Luo.


Neuron | 2015

Epigenomic Signatures of Neuronal Diversity in the Mammalian Brain

Alisa Mo; Eran A. Mukamel; Fred P. Davis; Chongyuan Luo; Gilbert L. Henry; Serge Picard; Mark A. Urich; Joseph R. Nery; Terrence J. Sejnowski; Ryan Lister; Sean R. Eddy; Joseph R. Ecker; Jeremy Nathans

Neuronal diversity is essential for mammalian brain function but poses a challenge to molecular profiling. To address the need for tools that facilitate cell-type-specific epigenomic studies, we developed the first affinity purification approach to isolate nuclei from genetically defined cell types in a mammal. We combine this technique with next-generation sequencing to show that three subtypes of neocortical neurons have highly distinctive epigenomic landscapes. Over 200,000 regions differ in chromatin accessibility and DNA methylation signatures characteristic of gene regulatory regions. By footprinting and motif analyses, these regions are predicted to bind distinct cohorts of neuron subtype-specific transcription factors. Neuronal epigenomes reflect both past and present gene expression, with DNA hyper-methylation at developmentally critical genes appearing as a novel epigenomic signature in mature neurons. Taken together, our findings link the functional and transcriptional complexity of neurons to their underlying epigenomic diversity.


Nature Methods | 2016

Phased diploid genome assembly with single-molecule real-time sequencing

Chen-Shan Chin; Paul Peluso; Fritz J. Sedlazeck; Maria Nattestad; Gregory T Concepcion; Alicia Clum; Christopher P. Dunn; Ronan O'Malley; Rosa Figueroa-Balderas; Abraham Morales-Cruz; Grant R. Cramer; Massimo Delledonne; Chongyuan Luo; Joseph R. Ecker; Dario Cantu; David Rank; Michael C. Schatz

While genome assembly projects have been successful in many haploid and inbred species, the assembly of noninbred or rearranged heterozygous genomes remains a major challenge. To address this challenge, we introduce the open-source FALCON and FALCON-Unzip algorithms (https://github.com/PacificBiosciences/FALCON/) to assemble long-read sequencing data into highly accurate, contiguous, and correctly phased diploid genomes. We generate new reference sequences for heterozygous samples including an F1 hybrid of Arabidopsis thaliana, the widely cultivated Vitis vinifera cv. Cabernet Sauvignon, and the coral fungus Clavicorona pyxidata, samples that have challenged short-read assembly approaches. The FALCON-based assemblies are substantially more contiguous and complete than alternate short- or long-read approaches. The phased diploid assembly enabled the study of haplotype structure and heterozygosities between homologous chromosomes, including the identification of widespread heterozygous structural variation within coding sequences.


Nature | 2015

An alternative pluripotent state confers interspecies chimaeric competency

Jun Wu; Daiji Okamura; Mo Li; Keiichiro Suzuki; Chongyuan Luo; Li Ma; Yupeng He; Zhongwei Li; Christopher Benner; Isao Tamura; Marie N. Krause; Joseph R. Nery; Tingting Du; Zhuzhu Zhang; Tomoaki Hishida; Yuta Takahashi; Emi Aizawa; Na Young Kim; Jeronimo Lajara; Pedro Guillen; Josep M. Campistol; Concepcion Rodriguez Esteban; Pablo J. Ross; Alan Saghatelian; Bing Ren; Joseph R. Ecker; Juan Carlos Izpisua Belmonte

Pluripotency, the ability to generate any cell type of the body, is an evanescent attribute of embryonic cells. Transitory pluripotent cells can be captured at different time points during embryogenesis and maintained as embryonic stem cells or epiblast stem cells in culture. Since ontogenesis is a dynamic process in both space and time, it seems counterintuitive that these two temporal states represent the full spectrum of organismal pluripotency. Here we show that by modulating culture parameters, a stem-cell type with unique spatial characteristics and distinct molecular and functional features, designated as region-selective pluripotent stem cells (rsPSCs), can be efficiently obtained from mouse embryos and primate pluripotent stem cells, including humans. The ease of culturing and editing the genome of human rsPSCs offers advantages for regenerative medicine applications. The unique ability of human rsPSCs to generate post-implantation interspecies chimaeric embryos may facilitate our understanding of early human development and evolution.


eLife | 2016

Epigenomic landscapes of retinal rods and cones

Alisa Mo; Chongyuan Luo; Fred P. Davis; Eran A. Mukamel; Gilbert L. Henry; Joseph R. Nery; Mark A. Urich; Serge Picard; Ryan Lister; Sean R. Eddy; Michael Beer; Joseph R. Ecker; Jeremy Nathans

Rod and cone photoreceptors are highly similar in many respects but they have important functional and molecular differences. Here, we investigate genome-wide patterns of DNA methylation and chromatin accessibility in mouse rods and cones and correlate differences in these features with gene expression, histone marks, transcription factor binding, and DNA sequence motifs. Loss of NR2E3 in rods shifts their epigenomes to a more cone-like state. The data further reveal wide differences in DNA methylation between retinal photoreceptors and brain neurons. Surprisingly, we also find a substantial fraction of DNA hypo-methylated regions in adult rods that are not in active chromatin. Many of these regions exhibit hallmarks of regulatory regions that were active earlier in neuronal development, suggesting that these regions could remain undermethylated due to the highly compact chromatin in mature rods. This work defines the epigenomic landscapes of rods and cones, revealing features relevant to photoreceptor development and function. DOI: http://dx.doi.org/10.7554/eLife.11613.001


Science | 2017

Single-cell methylomes identify neuronal subtypes and regulatory elements in mammalian cortex

Chongyuan Luo; Christopher L. Keown; Laurie Kurihara; Jingtian Zhou; Yupeng He; Junhao Li; Rosa Castanon; Jacinta Lucero; Joseph R. Nery; Justin P. Sandoval; Brian Bui; Terrence J. Sejnowski; Timothy T. Harkins; Eran A. Mukamel; M. Margarita Behrens; Joseph R. Ecker

Methylation and the single neuronal cell The presence or absence of methylation on chromosomal DNA can drive or repress gene expression. Now, a comprehensive map of methylation variation in neuronal cell populations, including a between-species comparison, illustrates how epigenetic diversity plays important roles in neuronal development. Luo et al. examined how DNA methylation is both similar and different within neurons at the single-nucleus level in humans and mice. They identified 16 mouse and 21 human neuronal clusters, with greater complexity of excitatory neurons in deep brain layers than in superficial layers. Science, this issue p. 600 Single-nucleus methylomes distinguish neuron types and predict conserved gene regulatory elements in mice and humans. The mammalian brain contains diverse neuronal types, yet we lack single-cell epigenomic assays that are able to identify and characterize them. DNA methylation is a stable epigenetic mark that distinguishes cell types and marks regulatory elements. We generated >6000 methylomes from single neuronal nuclei and used them to identify 16 mouse and 21 human neuronal subpopulations in the frontal cortex. CG and non-CG methylation exhibited cell type–specific distributions, and we identified regulatory elements with differential methylation across neuron types. Methylation signatures identified a layer 6 excitatory neuron subtype and a unique human parvalbumin-expressing inhibitory neuron subtype. We observed stronger cross-species conservation of regulatory elements in inhibitory neurons than in excitatory neurons. Single-nucleus methylomes expand the atlas of brain cell types and identify regulatory elements that drive conserved brain cell diversity.


Science | 2015

Exceptional epigenetics in the brain

Chongyuan Luo; Joseph R. Ecker

Non-CG DNA methylation modulates gene expression in the adult brain Cytosine (C) methylation, or mC, is a modification of DNA that regulates gene expression in various contexts such as development, cancer, and imprinting. In most mammalian somatic tissues, mC arises only when C is in a dinucleotide context followed by the nucleotide guanine (G), and the vast majority of these sites are methylated (mCG). However, in the adult mammalian brain, noncanonical cytosine methylation in a non-CG context occurs at a high level [mCH; H = adenine (A), C, or thymine (T)]. The quantity of non-CG methylation is inversely correlated with gene expression, but the mechanism by which mCH controls transcription has not been clear. Two recent studies (1, 2) suggest a mechanistic link between mCH and gene expression and substantially revise our view of gene regulation by DNA methylation in the brain, which was previously formed only on the basis of studies of mCG.


bioRxiv | 2017

Spatiotemporal DNA Methylome Dynamics of the Developing Mammalian Fetus

Yupeng He; Manoj Hariharan; David U. Gorkin; Diane E. Dickel; Chongyuan Luo; Rosa Castanon; Joseph R. Nery; Ah Young Lee; Brian A. Williams; Diane Trout; Henry Amrhein; Rongxin Fang; Huaming Chen; Bin Li; Axel Visel; Len A. Pennacchio; Bing Ren; Joseph R. Ecker

Genetic studies have revealed an essential role for cytosine DNA methylation in mammalian development. However, its spatiotemporal distribution in the developing embryo remains obscure. Here, we profiled the methylome landscapes of 12 mouse tissues/organs at 8 developmental stages spanning from early embryogenesis to birth. Indepth analysis of these spatiotemporal epigenome maps systematically delineated ~2 million methylation variant regions and uncovered widespread methylation dynamics at nearly one-half million tissue-specific enhancers, whose human counterparts were enriched for variants involved in genetic diseases. Strikingly, these predicted regulatory elements predominantly lose CG methylation during fetal development, whereas the trend is reversed after birth. Accumulation of non-CG methylation within gene bodies of key developmental transcription factors coincided with their transcriptional repression during later stages of fetal development. These spatiotemporal epigenomic maps provide a valuable resource for studying gene regulation during mammalian tissue/organ progression and for pinpointing regulatory elements involved in human developmental diseases.


bioRxiv | 2018

Global DNA methylation remodeling during direct reprogramming of fibroblasts to neurons

Chongyuan Luo; Qian Yi Lee; Orly L. Wapinski; Rosa Castanon; Joseph R. Nery; Sean M. Cullen; Margaret A. Goodell; Howard Y. Chang; Marius Wernig; Joseph R. Ecker

Direct reprogramming of fibroblasts to neurons induces widespread cellular and transcriptional reconfiguration. In this study, we characterized global epigenomic changes during direct reprogramming using whole-genome base-resolution DNA methylome (mC) sequencing. We found that the pioneer transcription factor Ascl1 alone is sufficient for inducing the uniquely neuronal feature of non-CG methylation (mCH), but co-expression of Brn2 and Mytl1 was required to establish a global mCH pattern reminiscent of mature cortical neurons. Ascl1 alone induced strong promoter CG methylation (mCG) of fibroblast specific genes, while BAM overexpression additionally targets a competing myogenic program and directs a more faithful conversion to neuronal cells. Ascl1 induces local demethylation at its binding sites. Surprisingly, co-expression with Brn2 and Mytl1 inhibited the ability of Ascl1 to induce demethylation, suggesting a contextual regulation of transcription factor - epigenome interaction. Finally, we found that de novo methylation by DNMT3A is required for efficient neuronal reprogramming.


Science | 2018

Dynamic DNA methylation: In the right place at the right time

Chongyuan Luo; Petra Hajkova; Joseph R. Ecker

The classical model of cytosine DNA methylation (the presence of 5-methylcytosine, 5mC) regulation depicts this covalent modification as a stable repressive regulator of promoter activity. However, whole-genome analysis of 5mC reveals widespread tissue- and cell type–specific patterns and pervasive dynamics during mammalian development. Here we review recent findings that delineate 5mC functions in developmental stages and diverse genomic compartments as well as discuss the molecular mechanisms that connect transcriptional regulation and 5mC. Beyond the newly appreciated dynamics, regulatory roles for 5mC have been suggested in new biological contexts, such as learning and memory or aging. The use of new single-cell measurement techniques and precise editing tools will enable functional analyses of 5mC in gene expression, clarifying its role in various biological processes.


eLife | 2018

Transcriptional and epigenomic landscapes of CNS and non-CNS vascular endothelial cells.

Mark F Sabbagh; Jacob S Heng; Chongyuan Luo; Rosa Castanon; Joseph R. Nery; Amir Rattner; Loyal A. Goff; Joseph R. Ecker; Jeremy Nathans

Vascular endothelial cell (EC) function depends on appropriate organ-specific molecular and cellular specializations. To explore genomic mechanisms that control this specialization, we have analyzed and compared the transcriptome, accessible chromatin, and DNA methylome landscapes from mouse brain, liver, lung, and kidney ECs. Analysis of transcription factor (TF) gene expression and TF motifs at candidate cis-regulatory elements reveals both shared and organ-specific EC regulatory networks. In the embryo, only those ECs that are adjacent to or within the central nervous system (CNS) exhibit canonical Wnt signaling, which correlates precisely with blood-brain barrier (BBB) differentiation and Zic3 expression. In the early postnatal brain, single-cell RNA-seq of purified ECs reveals (1) close relationships between veins and mitotic cells and between arteries and tip cells, (2) a division of capillary ECs into vein-like and artery-like classes, and (3) new endothelial subtype markers, including new validated tip cell markers.

Collaboration


Dive into the Chongyuan Luo's collaboration.

Top Co-Authors

Avatar

Joseph R. Ecker

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Joseph R. Nery

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Rosa Castanon

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy Nathans

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yupeng He

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Alisa Mo

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Angeline Rivkin

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Brian Bui

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Fred P. Davis

Howard Hughes Medical Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge