Network


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

Hotspot


Dive into the research topics where Paul G. Giresi is active.

Publication


Featured researches published by Paul G. Giresi.


Nature Methods | 2013

Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position

Jason D. Buenrostro; Paul G. Giresi; Lisa C. Zaba; Howard Y. Chang; William J. Greenleaf

We describe an assay for transposase-accessible chromatin using sequencing (ATAC-seq), based on direct in vitro transposition of sequencing adaptors into native chromatin, as a rapid and sensitive method for integrative epigenomic analysis. ATAC-seq captures open chromatin sites using a simple two-step protocol with 500–50,000 cells and reveals the interplay between genomic locations of open chromatin, DNA-binding proteins, individual nucleosomes and chromatin compaction at nucleotide resolution. We discovered classes of DNA-binding factors that strictly avoided, could tolerate or tended to overlap with nucleosomes. Using ATAC-seq maps of human CD4+ T cells from a proband obtained on consecutive days, we demonstrated the feasibility of analyzing an individuals epigenome on a timescale compatible with clinical decision-making.


Nature Genetics | 2010

A map of open chromatin in human pancreatic islets

Kyle J. Gaulton; Takao Nammo; Lorenzo Pasquali; Jeremy M. Simon; Paul G. Giresi; Marie P. Fogarty; Tami M. Panhuis; Piotr A. Mieczkowski; Antonio Secchi; Domenico Bosco; Thierry Berney; Eduard Montanya; Karen L. Mohlke; Jason D. Lieb; Jorge Ferrer

Tissue-specific transcriptional regulation is central to human disease. To identify regulatory DNA active in human pancreatic islets, we profiled chromatin by formaldehyde-assisted isolation of regulatory elements coupled with high-throughput sequencing (FAIRE-seq). We identified ∼80,000 open chromatin sites. Comparison of FAIRE-seq data from islets to that from five non-islet cell lines revealed ∼3,300 physically linked clusters of islet-selective open chromatin sites, which typically encompassed single genes that have islet-specific expression. We mapped sequence variants to open chromatin sites and found that rs7903146, a TCF7L2 intronic variant strongly associated with type 2 diabetes, is located in islet-selective open chromatin. We found that human islet samples heterozygous for rs7903146 showed allelic imbalance in islet FAIRE signals and that the variant alters enhancer activity, indicating that genetic variation at this locus acts in cis with local chromatin and regulatory changes. These findings illuminate the tissue-specific organization of cis-regulatory elements and show that FAIRE-seq can guide the identification of regulatory variants underlying disease susceptibility.


Molecular Cell | 2009

Highly Transcribed RNA Polymerase II Genes Are Impediments to Replication Fork Progression in Saccharomyces cerevisiae

Anna Azvolinsky; Paul G. Giresi; Jason D. Lieb; Virginia A. Zakian

Replication forks face multiple obstacles that slow their progression. By two-dimensional gel analysis, yeast forks pause at stable DNA protein complexes, and this pausing is greatly increased in the absence of the Rrm3 helicase. We used a genome-wide approach to identify 96 sites of very high DNA polymerase binding in wild-type cells. Most of these binding sites were not previously identified pause sites. Rather, the most highly represented genomic category among high DNA polymerase binding sites was the open reading frames (ORFs) of highly transcribed RNA polymerase II genes. Twice as many pause sites were identified in rrm3 compared with wild-type cells, as pausing in this strain occurred at both highly transcribed RNA polymerase II genes and the previously identified protein DNA complexes. ORFs of highly transcribed RNA polymerase II genes are a class of natural pause sites that are not exacerbated in rrm3 cells.


Nature Protocols | 2012

Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA

Jeremy M. Simon; Paul G. Giresi; Ian J. Davis; Jason D. Lieb

Eviction or destabilization of nucleosomes from chromatin is a hallmark of functional regulatory elements in eukaryotic genomes. Historically identified by nuclease hypersensitivity, these regulatory elements are typically bound by transcription factors or other regulatory proteins. FAIRE (formaldehyde-assisted isolation of regulatory elements) is an alternative approach to identify these genomic regions and has proven successful in a multitude of eukaryotic cell and tissue types. Cells or dissociated tissues are cross-linked briefly with formaldehyde, lysed and sonicated. Sheared chromatin is subjected to phenol/chloroform extraction and the isolated DNA, typically encompassing 1–3% of the human genome, is purified. We provide guidelines for quantitative analysis by PCR, microarrays or next-generation sequencing. Regulatory elements enriched by FAIRE have high concordance with those identified by nuclease hypersensitivity or chromatin immunoprecipitation (ChIP), and the entire procedure can be completed in 3 d. FAIRE has low technical variability, which allows its usage in large-scale studies of chromatin from normal or diseased tissues.


Genome Biology | 2011

ZINBA integrates local covariates with DNA-seq data to identify broad and narrow regions of enrichment, even within amplified genomic regions

Naim Rashid; Paul G. Giresi; Joseph G. Ibrahim; Wei Sun; Jason D. Lieb

ZINBA (Zero-Inflated Negative Binomial Algorithm) identifies genomic regions enriched in a variety of ChIP-seq and related next-generation sequencing experiments (DNA-seq), calling both broad and narrow modes of enrichment across a range of signal-to-noise ratios. ZINBA models and accounts for factors that co-vary with background or experimental signal, such as G/C content, and identifies enrichment in genomes with complex local copy number variations. ZINBA provides a single unified framework for analyzing DNA-seq experiments in challenging genomic contexts.Software website: http://code.google.com/p/zinba/


Nature Genetics | 2007

X-chromosome repression by localization of the C. elegans dosage compensation machinery to sites of transcription initiation

Sevinc Ercan; Paul G. Giresi; Christina M. Whittle; Xinmin Zhang; Roland D. Green; Jason D. Lieb

Among organisms with chromosome-based mechanisms of sex determination, failure to equalize expression of X-linked genes between the sexes is typically lethal. In C. elegans, XX hermaphrodites halve transcription from each X chromosome to match the output of XO males. Here, we mapped the binding location of the condensin homolog DPY-27 and the zinc finger protein SDC-3, two components of the C. elegans dosage compensation complex (DCC). We observed strong foci of DCC binding on X, surrounded by broader regions of localization. Binding foci, but not adjacent regions of localization, were distinguished by clusters of a 10-bp DNA motif, suggesting a recruitment-and-spreading mechanism for X recognition. The DCC was preferentially bound upstream of genes, suggesting modulation of transcriptional initiation and polymerase-coupled spreading. Stronger DCC binding upstream of genes with high transcriptional activity indicated a mechanism for tuning DCC activity at specific loci. These data aid in understanding how proteins involved in higher-order chromosome dynamics can regulate transcription at individual loci.


Current protocols in molecular biology | 2013

A Detailed Protocol for Formaldehyde‐Assisted Isolation of Regulatory Elements (FAIRE)

Jeremy M. Simon; Paul G. Giresi; Ian J. Davis; Jason D. Lieb

Nucleosome displacement is a key event in the regulation of gene expression in the eukaryotic genome. This unit details an approach called Formaldehyde‐Assisted Isolation of Regulatory Elements (FAIRE) for isolating nucleosome‐depleted regions. FAIRE does not rely on the use of antibodies or enzymes, and has proven successful in most eukaryotic cells and tissues. The set of regulatory elements enriched by FAIRE is similar to those identified through DNase hypersensitivity. The enriched fragments can be detected by quantitative PCR, tiling DNA microarrays, or next‐generation sequencing. Although the signal‐to‐noise ratio is typically lower than that observed for DNase assays, FAIRE has high sample‐to‐sample reproducibility, requires very low amounts of input material, is inexpensive, is amenable to high‐throughput adaptations, and is a relatively simple procedure with a high rate of success, even for those without extensive experience in molecular biology protocols. Curr. Protoc. Mol. Biol. 102:21.26.1–21.26.15.


Epigenetics & Chromatin | 2015

STAT3 acts through pre-existing nucleosome-depleted regions bound by FOS during an epigenetic switch linking inflammation to cancer.

Joseph D. Fleming; Paul G. Giresi; Marianne Lindahl-Allen; Elsa Beyer Krall; Jason D. Lieb; Kevin Struhl

BackgroundTransient induction of the Src oncoprotein in a non-transformed breast cell line can initiate an epigenetic switch to a cancer cell via a positive feedback loop that involves activation of the signal transducer and activator of transcription 3 protein (STAT3) and NF-κB transcription factors.ResultsWe show that during the transformation process, nucleosome-depleted regions (defined by formaldehyde-assisted isolation of regulatory elements (FAIRE)) are largely unchanged and that both before and during transformation, STAT3 binds almost exclusively to previously open chromatin regions. Roughly, a third of the transformation-inducible genes require STAT3 for the induction. STAT3 and NF-κB appear to drive the regulation of different gene sets during the transformation process. Interestingly, STAT3 directly regulates the expression of NFKB1, which encodes a subunit of NF-κB, and IL6, a cytokine that stimulates STAT3 activity. Lastly, many STAT3 binding sites are also bound by FOS and the expression of several AP-1 factors is altered during transformation in a STAT3-dependent manner, suggesting that STAT3 may cooperate with AP-1 proteins.ConclusionsThese observations uncover additional complexities to the inflammatory feedback loop that are likely to contribute to the epigenetic switch. In addition, gene expression changes during transformation, whether driven by pre-existing or induced transcription factors, occur largely through pre-existing nucleosome-depleted regions.


Cancer Cell | 2017

Repression of Stress-Induced LINE-1 Expression Protects Cancer Cell Subpopulations from Lethal Drug Exposure

Gulfem D. Guler; Charles Tindell; Robert M. Pitti; Catherine Wilson; Katrina Nichols; Tommy K. Cheung; Hyo-Jin Kim; Matthew Wongchenko; Yibing Yan; Benjamin Haley; Trinna L. Cuellar; Joshua D. Webster; Navneet Alag; Ganapati V. Hegde; Erica Jackson; Tracy Leah Nance; Paul G. Giresi; Kuan-Bei Chen; Jinfeng Liu; Suchit Jhunjhunwala; Jeffrey Settleman; Jean-Philippe Stephan; David Arnott; Marie Classon

Maintenance of phenotypic heterogeneity within cell populations is an evolutionarily conserved mechanism that underlies population survival upon stressful exposures. We show that the genomes of a cancer cell subpopulation that survives treatment with otherwise lethal drugs, the drug-tolerant persisters (DTPs), exhibit a repressed chromatin state characterized by increased methylation of histone H3 lysines 9 and 27 (H3K9 and H3K27). We also show that survival of DTPs is, in part, maintained by regulators of H3K9me3-mediated heterochromatin formation and that the observed increase in H3K9me3 in DTPs is most prominent over long interspersed repeat element 1 (LINE-1). Disruption of the repressive chromatin over LINE-1 elements in DTPs results in DTP ablation, which is partially rescued by reducing LINE-1 expression or function.


Nature Methods | 2006

How to find an opening (or lots of them).

Paul G. Giresi; Jason D. Lieb

DNase-chip and DNase-array: similar names for two different new approaches that give a genomic perspective to the conventional DNase I hypersensitivity assay used to measure chromatin accessibility.

Collaboration


Dive into the Paul G. Giresi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy M. Simon

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Kun Qu

Stanford University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian J. Davis

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Rui Li

Stanford University

View shared research outputs
Top Co-Authors

Avatar

Ryan M. McDaniell

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge