Network


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

Hotspot


Dive into the research topics where Lovelace J. Luquette is active.

Publication


Featured researches published by Lovelace J. Luquette.


Nature | 2011

Comprehensive analysis of the chromatin landscape in Drosophila melanogaster

Peter V. Kharchenko; Artyom A. Alekseyenko; Yuri B. Schwartz; Aki Minoda; Nicole C. Riddle; Jason Ernst; Peter J. Sabo; Erica Larschan; Andrey A. Gorchakov; Tingting Gu; Daniela Linder-Basso; Annette Plachetka; Gregory Shanower; Michael Y. Tolstorukov; Lovelace J. Luquette; Ruibin Xi; Youngsook L. Jung; Richard Park; Eric P. Bishop; Theresa P. Canfield; Richard Sandstrom; Robert E. Thurman; David M. MacAlpine; John A. Stamatoyannopoulos; Manolis Kellis; Sarah C. R. Elgin; Mitzi I. Kuroda; Vincenzo Pirrotta; Gary H. Karpen; Peter J. Park

Chromatin is composed of DNA and a variety of modified histones and non-histone proteins, which have an impact on cell differentiation, gene regulation and other key cellular processes. Here we present a genome-wide chromatin landscape for Drosophila melanogaster based on eighteen histone modifications, summarized by nine prevalent combinatorial patterns. Integrative analysis with other data (non-histone chromatin proteins, DNase I hypersensitivity, GRO-Seq reads produced by engaged polymerase, short/long RNA products) reveals discrete characteristics of chromosomes, genes, regulatory elements and other functional domains. We find that active genes display distinct chromatin signatures that are correlated with disparate gene lengths, exon patterns, regulatory functions and genomic contexts. We also demonstrate a diversity of signatures among Polycomb targets that include a subset with paused polymerase. This systematic profiling and integrative analysis of chromatin signatures provides insights into how genomic elements are regulated, and will serve as a resource for future experimental investigations of genome structure and function.


Science | 2012

Landscape of somatic retrotransposition in human cancers.

Eunjung Lee; Rebecca Iskow; Lixing Yang; Omer Gokcumen; Psalm Haseley; Lovelace J. Luquette; Jens Lohr; Christopher C. Harris; Li Ding; Richard Wilson; David A. Wheeler; Richard A. Gibbs; Raju Kucherlapati; Charles Lee; Peter V. Kharchenko; Peter J. Park

Movement in the Cancer Genome Transposable elements are genetic sequences that can replicate and move within the genome. The factors that make an element mobile are unknown but are generally considered rare in mammals. Lee et al. (p. 967, published online 28 June) analyzed five cancer types occurring among several individuals and found that three types of epithelial tumors exhibited high rates of element movement relative to brain and blood cancers. Furthermore, these somatically acquired, tumor-specific elements targeted genes in colorectal cancer that, when disrupted, impact gene expression and thus may be a factor in the progression of the cancers. Whole-genome sequencing provides evidence for somatic insertions in colorectal, prostate, and ovarian cancers. Transposable elements (TEs) are abundant in the human genome, and some are capable of generating new insertions through RNA intermediates. In cancer, the disruption of cellular mechanisms that normally suppress TE activity may facilitate mutagenic retrotranspositions. We performed single-nucleotide resolution analysis of TE insertions in 43 high-coverage whole-genome sequencing data sets from five cancer types. We identified 194 high-confidence somatic TE insertions, as well as thousands of polymorphic TE insertions in matched normal genomes. Somatic insertions were present in epithelial tumors but not in blood or brain cancers. Somatic L1 insertions tend to occur in genes that are commonly mutated in cancer, disrupt the expression of the target genes, and are biased toward regions of cancer-specific DNA hypomethylation, highlighting their potential impact in tumorigenesis.


Science | 2015

Somatic mutation in single human neurons tracks developmental and transcriptional history

Michael A. Lodato; Mollie B. Woodworth; Semin Lee; Gilad D. Evrony; Bhaven K. Mehta; Amir Karger; Soohyun Lee; Thomas Chittenden; Alissa M. D’Gama; Xuyu Cai; Lovelace J. Luquette; Eunjung Lee; Peter J. Park; Christopher A. Walsh

Individualized neuronal mutations in the human brain The neurons of the human brain can last for decades, carrying out computational and signaling functions. Lodato et al. analyzed the DNA of individual neurons sampled from postmortem human brains and found that individual neurons acquired somatic mutations (see the Perspective by Linnarsson). The mechanism of mutation involved gene transcription rather than DNA replication. Thus, postmitotic neurons would seem to be their own worst enemy: Genes used for neuronal function are the very genes put most at risk of somatic mutation. Science, this issue p. 94; see also p. 37 Human brains are built from intermingled clones of cells that carry mutations linked to their use of particular neuronal genes. [Also see Perspective by Linnarsson] Neurons live for decades in a postmitotic state, their genomes susceptible to DNA damage. Here we survey the landscape of somatic single-nucleotide variants (SNVs) in the human brain. We identified thousands of somatic SNVs by single-cell sequencing of 36 neurons from the cerebral cortex of three normal individuals. Unlike germline and cancer SNVs, which are often caused by errors in DNA replication, neuronal mutations appear to reflect damage during active transcription. Somatic mutations create nested lineage trees, allowing them to be dated relative to developmental landmarks and revealing a polyclonal architecture of the human cerebral cortex. Thus, somatic mutations in the brain represent a durable and ongoing record of neuronal life history, from development through postmitotic function.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Copy number variation detection in whole-genome sequencing data using the Bayesian information criterion

Ruibin Xi; Angela Hadjipanayis; Lovelace J. Luquette; Tae-Min Kim; Eunjung Lee; Jianhua Zhang; Mark D. Johnson; Donna M. Muzny; David A. Wheeler; Richard A. Gibbs; Raju Kucherlapati; Peter J. Park

DNA copy number variations (CNVs) play an important role in the pathogenesis and progression of cancer and confer susceptibility to a variety of human disorders. Array comparative genomic hybridization has been used widely to identify CNVs genome wide, but the next-generation sequencing technology provides an opportunity to characterize CNVs genome wide with unprecedented resolution. In this study, we developed an algorithm to detect CNVs from whole-genome sequencing data and applied it to a newly sequenced glioblastoma genome with a matched control. This read-depth algorithm, called BIC-seq, can accurately and efficiently identify CNVs via minimizing the Bayesian information criterion. Using BIC-seq, we identified hundreds of CNVs as small as 40 bp in the cancer genome sequenced at 10× coverage, whereas we could only detect large CNVs (> 15 kb) in the array comparative genomic hybridization profiles for the same genome. Eighty percent (14/16) of the small variants tested (110 bp to 14 kb) were experimentally validated by quantitative PCR, demonstrating high sensitivity and true positive rate of the algorithm. We also extended the algorithm to detect recurrent CNVs in multiple samples as well as deriving error bars for breakpoints using a Gibbs sampling approach. We propose this statistical approach as a principled yet practical and efficient method to estimate CNVs in whole-genome sequencing data.


Cell | 2013

Diverse Mechanisms of Somatic Structural Variations in Human Cancer Genomes

Lixing Yang; Lovelace J. Luquette; Nils Gehlenborg; Ruibin Xi; Psalm Haseley; Chih Heng Hsieh; Chengsheng Zhang; Xiaojia Ren; Alexei Protopopov; Lynda Chin; Raju Kucherlapati; Charles Lee; Peter J. Park

Identification of somatic rearrangements in cancer genomes has accelerated through analysis of high-throughput sequencing data. However, characterization of complex structural alterations and their underlying mechanisms remains inadequate. Here, applying an algorithm to predict structural variations from short reads, we report a comprehensive catalog of somatic structural variations and the mechanisms generating them, using high-coverage whole-genome sequencing data from 140 patients across ten tumor types. We characterize the relative contributions of different types of rearrangements and their mutational mechanisms, find that ∼20% of the somatic deletions are complex deletions formed by replication errors, and describe the differences between the mutational mechanisms in somatic and germline alterations. Importantly, we provide detailed reconstructions of the events responsible for loss of CDKN2A/B and gain of EGFR in glioblastoma, revealing that these alterations can result from multiple mechanisms even in a single genome and that both DNA double-strand breaks and replication errors drive somatic rearrangements.Identification of somatic rearrangements in cancer genomes has accelerated through analysis of high-throughput sequencing data. However, characterization of complex structural alterations and their underlying mechanisms remains inadequate. Here, applying an algorithm to predict structural variations from short reads, we report a comprehensive catalog of somatic structural variations and the mechanisms generating them, using high-coverage whole-genome sequencing data from 140 patients across ten tumor types. We characterize the relative contributions of different types of rearrangements and their mutational mechanisms, find that ~20% of the somatic deletions are complex deletions formed by replication errors, and describe the differences between the mutational mechanisms in somatic and germline alterations. Importantly, we provide detailed reconstructions of the events responsible for loss of CDKN2A/B and gain of EGFR in glioblastoma, revealing that these alterations can result from multiple mechanisms even in a single genome and that both DNA double-strand breaks and replication errors drive somatic rearrangements.


Genome Biology | 2010

Estimating Enrichment of Repetitive Elements from High-throughput Sequence Data

Daniel S. Day; Lovelace J. Luquette; Peter J. Park; Peter V. Kharchenko

We describe computational methods for analysis of repetitive elements from short-read sequencing data, and apply them to study histone modifications associated with the repetitive elements in human and mouse cells. Our results demonstrate that while accurate enrichment estimates can be obtained for individual repeat types and small sets of repeat instances, there are distinct combinatorial patterns of chromatin marks associated with major annotated repeat families, including H3K27me3/H3K9me3 differences among the endogenous retroviral element classes.


Nature Biotechnology | 2012

Systematic identification of synergistic drug pairs targeting HIV

Xu Tan; Long Hu; Lovelace J. Luquette; Geng Gao; Yifang Liu; Hongjing Qu; Ruibin Xi; Zhi John Lu; Peter J. Park; Stephen J. Elledge

The systematic identification of effective drug combinations has been hindered by the unavailability of methods that can explore the large combinatorial search space of drug interactions. Here we present multiplex screening for interacting compounds (MuSIC), which expedites the comprehensive assessment of pairwise compound interactions. We examined ∼500,000 drug pairs from 1,000 US Food and Drug Administration (FDA)-approved or clinically tested drugs and identified drugs that synergize to inhibit HIV replication. Our analysis reveals an enrichment of anti-inflammatory drugs in drug combinations that synergize against HIV. As inflammation accompanies HIV infection, these findings indicate that inhibiting inflammation could curb HIV propagation. Multiple drug pairs identified in this study, including various glucocorticoids and nitazoxanide (NTZ), synergize by targeting different steps in the HIV life cycle. MuSIC can be applied to a wide variety of disease-relevant screens to facilitate efficient identification of compound combinations.


BMC Bioinformatics | 2010

rSW-seq: Algorithm for detection of copy number alterations in deep sequencing data

Tae-Min Kim; Lovelace J. Luquette; Ruibin Xi; Peter J. Park

BackgroundRecent advances in sequencing technologies have enabled generation of large-scale genome sequencing data. These data can be used to characterize a variety of genomic features, including the DNA copy number profile of a cancer genome. A robust and reliable method for screening chromosomal alterations would allow a detailed characterization of the cancer genome with unprecedented accuracy.ResultsWe develop a method for identification of copy number alterations in a tumor genome compared to its matched control, based on application of Smith-Waterman algorithm to single-end sequencing data. In a performance test with simulated data, our algorithm shows >90% sensitivity and >90% precision in detecting a single copy number change that contains approximately 500 reads for the normal sample. With 100-bp reads, this corresponds to a ~50 kb region for 1X genome coverage of the human genome. We further refine the algorithm to develop rSW-seq, (recursive Smith-Waterman-seq) to identify alterations in a complex configuration, which are commonly observed in the human cancer genome. To validate our approach, we compare our algorithm with an existing algorithm using simulated and publicly available datasets. We also compare the sequencing-based profiles to microarray-based results.ConclusionWe propose rSW-seq as an efficient method for detecting copy number changes in the tumor genome.


Nucleic Acids Research | 2014

Impact of sequencing depth in ChIP-seq experiments

Youngsook L. Jung; Lovelace J. Luquette; Joshua W. K. Ho; Francesco Ferrari; Michael Y. Tolstorukov; Aki Minoda; Robbyn Issner; Charles B. Epstein; Gary H. Karpen; Mitzi I. Kuroda; Peter J. Park

In a chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) experiment, an important consideration in experimental design is the minimum number of sequenced reads required to obtain statistically significant results. We present an extensive evaluation of the impact of sequencing depth on identification of enriched regions for key histone modifications (H3K4me3, H3K36me3, H3K27me3 and H3K9me2/me3) using deep-sequenced datasets in human and fly. We propose to define sufficient sequencing depth as the number of reads at which detected enrichment regions increase <1% for an additional million reads. Although the required depth depends on the nature of the mark and the state of the cell in each experiment, we observe that sufficient depth is often reached at <20 million reads for fly. For human, there are no clear saturation points for the examined datasets, but our analysis suggests 40–50 million reads as a practical minimum for most marks. We also devise a mathematical model to estimate the sufficient depth and total genomic coverage of a mark. Lastly, we find that the five algorithms tested do not agree well for broad enrichment profiles, especially at lower depths. Our findings suggest that sufficient sequencing depth and an appropriate peak-calling algorithm are essential for ensuring robustness of conclusions derived from ChIP-seq data.


European Journal of Medical Genetics | 2013

Mutation of KCNJ8 in a patient with Cantú syndrome with unique vascular abnormalities - support for the role of K(ATP) channels in this condition.

Catherine A. Brownstein; Meghan C. Towne; Lovelace J. Luquette; David J. Harris; Nicholas S. Marinakis; Peter Meinecke; Kerstin Kutsche; Philippe M. Campeau; David M. Margulies; Pankaj B. Agrawal; Alan H. Beggs

KCNJ8 (NM_004982) encodes the pore forming subunit of one of the ATP-sensitive inwardly rectifying potassium (KATP) channels. KCNJ8 sequence variations are traditionally associated with J-wave syndromes, involving ventricular fibrillation and sudden cardiac death. Recently, the KATP gene ABCC9 (SUR2, NM_020297) has been associated with the multi-organ disorder Cantú syndrome or hypertrichotic osteochondrodysplasia (MIM 239850) (hypertrichosis, macrosomia, osteochondrodysplasia, and cardiomegaly). Here, we report on a patient with a de novo nonsynonymous KCNJ8 SNV (p.V65M) and Cantú syndrome, who tested negative for mutations in ABCC9. The genotype and multi-organ abnormalities of this patient are reviewed. A careful screening of the KATP genes should be performed in all individuals diagnosed with Cantú syndrome and no mutation in ABCC9.

Collaboration


Dive into the Lovelace J. Luquette's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexei Protopopov

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Charles Lee

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lynda Chin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge