Network


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

Hotspot


Dive into the research topics where John Jack is active.

Publication


Featured researches published by John Jack.


Genome Research | 2014

Natural variation in genome architecture among 205 Drosophila melanogaster Genetic Reference Panel lines

Wen Huang; Andreas Massouras; Yutaka Inoue; Jason A. Peiffer; Miquel Ràmia; Aaron M. Tarone; Lavanya Turlapati; Thomas Zichner; Dianhui Zhu; Richard F. Lyman; Michael M. Magwire; Kerstin P. Blankenburg; Mary Anna Carbone; Kyle Chang; Lisa L. Ellis; Sonia Fernandez; Yi Han; Gareth Highnam; Carl E. Hjelmen; John Jack; Mehwish Javaid; Joy Jayaseelan; Divya Kalra; Sandy Lee; Lora Lewis; Mala Munidasa; Fiona Ongeri; Shohba Patel; Lora Perales; Agapito Perez

The Drosophila melanogaster Genetic Reference Panel (DGRP) is a community resource of 205 sequenced inbred lines, derived to improve our understanding of the effects of naturally occurring genetic variation on molecular and organismal phenotypes. We used an integrated genotyping strategy to identify 4,853,802 single nucleotide polymorphisms (SNPs) and 1,296,080 non-SNP variants. Our molecular population genomic analyses show higher deletion than insertion mutation rates and stronger purifying selection on deletions. Weaker selection on insertions than deletions is consistent with our observed distribution of genome size determined by flow cytometry, which is skewed toward larger genomes. Insertion/deletion and single nucleotide polymorphisms are positively correlated with each other and with local recombination, suggesting that their nonrandom distributions are due to hitchhiking and background selection. Our cytogenetic analysis identified 16 polymorphic inversions in the DGRP. Common inverted and standard karyotypes are genetically divergent and account for most of the variation in relatedness among the DGRP lines. Intriguingly, variation in genome size and many quantitative traits are significantly associated with inversions. Approximately 50% of the DGRP lines are infected with Wolbachia, and four lines have germline insertions of Wolbachia sequences, but effects of Wolbachia infection on quantitative traits are rarely significant. The DGRP complements ongoing efforts to functionally annotate the Drosophila genome. Indeed, 15% of all D. melanogaster genes segregate for potentially damaged proteins in the DGRP, and genome-wide analyses of quantitative traits identify novel candidate genes. The DGRP lines, sequence data, genotypes, quality scores, phenotypes, and analysis and visualization tools are publicly available.


Alzheimers & Dementia | 2017

Metabolic network failures in Alzheimer's disease—A biochemical road map

Jon B. Toledo; Matthias Arnold; Gabi Kastenmüller; Rui Chang; Rebecca A. Baillie; Xianlin Han; Madhav Thambisetty; Jessica D. Tenenbaum; Karsten Suhre; J. Will Thompson; Lisa St. John-Williams; Siamak MahmoudianDehkordi; Daniel M. Rotroff; John Jack; Alison A. Motsinger-Reif; Shannon L. Risacher; Colette Blach; Joseph E. Lucas; Tyler Massaro; Gregory Louie; Hongjie Zhu; Guido Dallmann; Kristaps Klavins; Therese Koal; Sungeun Kim; Kwangsik Nho; Li Shen; Ramon Casanova; Sudhir Varma; Cristina Legido-Quigley

The Alzheimers Disease Research Summits of 2012 and 2015 incorporated experts from academia, industry, and nonprofit organizations to develop new research directions to transform our understanding of Alzheimers disease (AD) and propel the development of critically needed therapies. In response to their recommendations, big data at multiple levels are being generated and integrated to study network failures in disease. We used metabolomics as a global biochemical approach to identify peripheral metabolic changes in AD patients and correlate them to cerebrospinal fluid pathology markers, imaging features, and cognitive performance.


Environmental Health Perspectives | 2015

Population-Based in Vitro Hazard and Concentration–Response Assessment of Chemicals: The 1000 Genomes High-Throughput Screening Study

Nour Abdo; Menghang Xia; Chad Brown; Oksana Kosyk; Ruili Huang; Srilatha Sakamuru; Yi Hui Zhou; John Jack; Paul J. Gallins; Kai Xia; Yun Li; Weihsueh A. Chiu; Alison A. Motsinger-Reif; Christopher P. Austin; Raymond R. Tice; Ivan Rusyn; Fred A. Wright

Background: Understanding of human variation in toxicity to environmental chemicals remains limited, so human health risk assessments still largely rely on a generic 10-fold factor (10½ each for toxicokinetics and toxicodynamics) to account for sensitive individuals or subpopulations. Objectives: We tested a hypothesis that population-wide in vitro cytotoxicity screening can rapidly inform both the magnitude of and molecular causes for interindividual toxicodynamic variability. Methods: We used 1,086 lymphoblastoid cell lines from the 1000 Genomes Project, representing nine populations from five continents, to assess variation in cytotoxic response to 179 chemicals. Analysis included assessments of population variation and heritability, and genome-wide association mapping, with attention to phenotypic relevance to human exposures. Results: For about half the tested compounds, cytotoxic response in the 1% most “sensitive” individual occurred at concentrations within a factor of 10½ (i.e., approximately 3) of that in the median individual; however, for some compounds, this factor was > 10. Genetic mapping suggested important roles for variation in membrane and transmembrane genes, with a number of chemicals showing association with SNP rs13120371 in the solute carrier SLC7A11, previously implicated in chemoresistance. Conclusions: This experimental approach fills critical gaps unaddressed by recent large-scale toxicity testing programs, providing quantitative, experimentally based estimates of human toxicodynamic variability, and also testable hypotheses about mechanisms contributing to interindividual variation. Citation: Abdo N, Xia M, Brown CC, Kosyk O, Huang R, Sakamuru S, Zhou YH, Jack JR, Gallins P, Xia K, Li Y, Chiu WA, Motsinger-Reif AA, Austin CP, Tice RR, Rusyn I, Wright FA. 2015. Population-based in vitro hazard and concentration–response assessment of chemicals: the 1000 Genomes high-throughput screening study. Environ Health Perspect 123:458–466; http://dx.doi.org/10.1289/ehp.1408775


Environmental Health Perspectives | 2015

Using ToxCast™ Data to Reconstruct Dynamic Cell State Trajectories and Estimate Toxicological Points of Departure

Imran Shah; R. Woodrow Setzer; John Jack; Keith A. Houck; Richard S. Judson; Thomas B. Knudsen; Jie Liu; Matthew T. Martin; David M. Reif; Ann M. Richard; Russell S. Thomas; Kevin M. Crofton; David J. Dix; Robert J. Kavlock

Background: High-content imaging (HCI) allows simultaneous measurement of multiple cellular phenotypic changes and is an important tool for evaluating the biological activity of chemicals. Objectives: Our goal was to analyze dynamic cellular changes using HCI to identify the “tipping point” at which the cells did not show recovery towards a normal phenotypic state. Methods: HCI was used to evaluate the effects of 967 chemicals (in concentrations ranging from 0.4 to 200 μM) on HepG2 cells over a 72-hr exposure period. The HCI end points included p53, c-Jun, histone H2A.x, α-tubulin, histone H3, alpha tubulin, mitochondrial membrane potential, mitochondrial mass, cell cycle arrest, nuclear size, and cell number. A computational model was developed to interpret HCI responses as cell-state trajectories. Results: Analysis of cell-state trajectories showed that 336 chemicals produced tipping points and that HepG2 cells were resilient to the effects of 334 chemicals up to the highest concentration (200 μM) and duration (72 hr) tested. Tipping points were identified as concentration-dependent transitions in system recovery, and the corresponding critical concentrations were generally between 5 and 15 times (25th and 75th percentiles, respectively) lower than the concentration that produced any significant effect on HepG2 cells. The remaining 297 chemicals require more data before they can be placed in either of these categories. Conclusions: These findings show the utility of HCI data for reconstructing cell state trajectories and provide insight into the adaptation and resilience of in vitro cellular systems based on tipping points. Cellular tipping points could be used to define a point of departure for risk-based prioritization of environmental chemicals. Citation: Shah I, Setzer RW, Jack J, Houck KA, Judson RS, Knudsen TB, Liu J, Martin MT, Reif DM, Richard AM, Thomas RS, Crofton KM, Dix DJ, Kavlock RJ. 2016. Using ToxCast™ data to reconstruct dynamic cell state trajectories and estimate toxicological points of departure. Environ Health Perspect 124:910–919; http://dx.doi.org/10.1289/ehp.1409029


Pharmacogenomics | 2014

Genome-wide association and pharmacological profiling of 29 anticancer agents using lymphoblastoid cell lines

Chad Brown; Tammy M. Havener; Marisa W. Medina; John Jack; Ronald M. Krauss; Howard L. McLeod; Alison A. Motsinger-Reif

AIM Association mapping with lymphoblastoid cell lines (LCLs) is a promising approach in pharmacogenomics research, and in the current study we utilized LCLs to perform association mapping for 29 chemotherapy drugs. MATERIALS & METHODS Currently, we use LCLs to perform genome-wide association mapping of the cytotoxic response of 520 European-Americans to 29 different anticancer drugs; the largest LCL study to date. A novel association approach using a multivariate analysis of covariance design was employed with the software program MAGWAS, testing for differences in the dose-response profiles between genotypes without making assumptions about the response curve or the biologic mode of association. Additionally, by classifying 25 of the 29 drugs into eight families according to structural and mechanistic relationships, MAGWAS was used to test for associations that were shared across each drug family. Finally, a unique algorithm using multivariate responses and multiple linear regressions across pairs of response curves was used for unsupervised clustering of drugs. RESULTS Among the single-drug studies, suggestive associations were obtained for 18 loci, 12 within/near genes. Three of these, MED12L, CHN2 and MGMT, have been previously implicated in cancer pharmacogenomics. The drug family associations resulted in four additional suggestive loci (three contained within/near genes). One of these genes, HDAC4, associated with the DNA alkylating agents, shows possible clinical interactions with temozolomide. For the drug clustering analysis, 18 of 25 drugs clustered into the appropriate family. CONCLUSION This study demonstrates the utility of LCLs in identifying genes that have clinical importance in drug response and for assigning unclassified agents to specific drug families, and proposes new candidate genes for follow-up in a large number of chemotherapy drugs.


Biodata Mining | 2014

An adaptive permutation approach for genome-wide association study: evaluation and recommendations for use

Ronglin Che; John Jack; Alison A. Motsinger-Reif; Chad Brown

BackgroundPermutation testing is a robust and popular approach for significance testing in genomic research, which has the broad advantage of estimating significance non-parametrically, thereby safe guarding against inflated type I error rates. However, the computational efficiency remains a challenging issue that limits its wide application, particularly in genome-wide association studies (GWAS). Because of this, adaptive permutation strategies can be employed to make permutation approaches feasible. While these approaches have been used in practice, there is little research into the statistical properties of these approaches, and little guidance into the proper application of such a strategy for accurate p-value estimation at the GWAS level.MethodsIn this work, we advocate an adaptive permutation procedure that is statistically valid as well as computationally feasible in GWAS. We perform extensive simulation experiments to evaluate the robustness of the approach to violations of modeling assumptions and compare the power of the adaptive approach versus standard approaches. We also evaluate the parameter choices in implementing the adaptive permutation approach to provide guidance on proper implementation in real studies. Additionally, we provide an example of the application of adaptive permutation testing on real data.ResultsThe results provide sufficient evidence that the adaptive test is robust to violations of modeling assumptions. In addition, even when modeling assumptions are correct, the power achieved by adaptive permutation is identical to the parametric approach over a range of significance thresholds and effect sizes under the alternative. A framework for proper implementation of the adaptive procedure is also generated.ConclusionsWhile the adaptive permutation approach presented here is not novel, the current study provides evidence of the validity of the approach, and importantly provides guidance on the proper implementation of such a strategy. Additionally, tools are made available to aid investigators in implementing these approaches.


Scientific Data | 2017

Targeted metabolomics and medication classification data from participants in the ADNI1 cohort

Lisa St. John-Williams; Colette Blach; Jon B. Toledo; Daniel M. Rotroff; Sungeun Kim; Kristaps Klavins; Rebecca A. Baillie; Xianlin Han; Siamak MahmoudianDehkordi; John Jack; Tyler Massaro; Joseph E. Lucas; Gregory Louie; Alison A. Motsinger-Reif; Shannon L. Risacher; Andrew J. Saykin; Gabi Kastenmüller; Matthias Arnold; Therese Koal; M. Arthur Moseley; Lara M. Mangravite; Mette A. Peters; Jessica D. Tenenbaum; J. Will Thompson; Rima Kaddurah-Daouk

Alzheimer’s disease (AD) is the most common neurodegenerative disease presenting major health and economic challenges that continue to grow. Mechanisms of disease are poorly understood but significant data point to metabolic defects that might contribute to disease pathogenesis. The Alzheimer Disease Metabolomics Consortium (ADMC) in partnership with Alzheimer Disease Neuroimaging Initiative (ADNI) is creating a comprehensive biochemical database for AD. Using targeted and non- targeted metabolomics and lipidomics platforms we are mapping metabolic pathway and network failures across the trajectory of disease. In this report we present quantitative metabolomics data generated on serum from 199 control, 356 mild cognitive impairment and 175 AD subjects enrolled in ADNI1 using AbsoluteIDQ-p180 platform, along with the pipeline for data preprocessing and medication classification for confound correction. The dataset presented here is the first of eight metabolomics datasets being generated for broad biochemical investigation of the AD metabolome. We expect that these collective metabolomics datasets will provide valuable resources for researchers to identify novel molecular mechanisms contributing to AD pathogenesis and disease phenotypes.


Pharmacogenetics and Genomics | 2016

A comparison of DMET Plus microarray and genome-wide technologies by assessing population substructure.

Jami N. Jackson; Kevin M. Long; Yijing He; Alison A. Motsinger-Reif; Howard L. McLeod; John Jack

Objective The capacity of the Affymetrix drug metabolism enzymes and transporters (DMET) Plus pharmacogenomics genotyping chip to estimate population substructure and cryptic relatedness was evaluated. The results were compared with estimates using genome-wide HapMap data for the same individuals. Methods For 301 unrelated individuals, spanning three continental populations and one admixed population, genotypic data were collected using the Affymetrix DMET Plus microarray. Genome-wide data on these individuals were obtained from HapMap release 3. Population substructure was assessed using Eigenstrat and ADMIXTURE software for both platforms. Cryptic relatedness was explored by inbreeding coefficient estimation. Nonparametric tests were used to determine correlations of the analytical results of the two genotyping platforms. Results Principal components analysis identified population substructure for both datasets, with 15.8 and 16.6% of the total variance explained in the first two principal components for DMET Plus and HapMap data, respectively. ADMIXTURE results correctly identified four subpopulations within each dataset. Nonparametric rank correlations indicated significant associations between analyses with an average &rgr;=0.7272 (P<10–16) across the three continental populations and &rgr;=0.4888 for the admixed population. Concordance correlation coefficients (average &rgr;c=0.9693 across all four subpopulations) strongly indicate concordance between ADMIXTURE results. Inbreeding coefficients were slightly inflated (16 individuals>0.15) using DMET Plus data and no cryptic relatedness was indicated using HapMap data. The inflated inbreeding estimation could be because of the limited number of markers provided by DMET as a random sample of 1832 markers from HapMap also yielded inflated estimates of cryptic relatedness (39 individuals>0.15). Furthermore, use of single nucleotide polymorphisms located in genes involved in metabolism and transport may have different allele frequencies in subpopulations than single nucleotide polymorphisms sampled from the whole genome. Conclusion The DMET Plus pharmacogenomics genotyping chip is effective in quantifying population substructure across the three continental populations and inferring the presence of an admixed population. On the basis of our results, these microarrays offer sufficient depth for covariate adjustment of population substructure in genomic association studies.


Clinical Pharmacology & Therapeutics | 2018

Genetic Variants in HSD17B3, SMAD3, and IPO11 Impact Circulating Lipids in Response to Fenofibrate in Individuals With Type 2 Diabetes

Daniel M. Rotroff; Sonja S. Pijut; Skylar W. Marvel; John Jack; Tammy M. Havener; Aurora Pujol; Agatha Schlüter; Gregory A. Graf; Henry N. Ginsberg; Hetal Shah; He Gao; Mario‐Luca Morieri; Alessandro Doria; Josyf C. Mychaleckyi; Howard L. McLeod; John B. Buse; Michael Wagner; Alison A. Motsinger-Reif

Individuals with type 2 diabetes (T2D) and dyslipidemia are at an increased risk of cardiovascular disease. Fibrates are a class of drugs prescribed to treat dyslipidemia, but variation in response has been observed. To evaluate common and rare genetic variants that impact lipid responses to fenofibrate in statin‐treated patients with T2D, we examined lipid changes in response to fenofibrate therapy using a genomewide association study (GWAS). Associations were followed‐up using gene expression studies in mice. Common variants in SMAD3 and IPO11 were marginally associated with lipid changes in black subjects (P < 5 × 10‐6). Rare variant and gene expression changes were assessed using a false discovery rate approach. AKR7A3 and HSD17B13 were associated with lipid changes in white subjects (q < 0.2). Mice fed fenofibrate displayed reductions in Hsd17b13 gene expression (q < 0.1). Associations of variants in SMAD3, IPO11, and HSD17B13, with gene expression changes in mice indicate that transforming growth factor‐beta (TGF‐β) and NRF2 signaling pathways may influence fenofibrate effects on dyslipidemia in patients with T2D.


Biodata Mining | 2014

PGxClean: a quality control GUI for the Affymetrix DMET chip and other candidate gene studies with non-biallelic alleles

Daniel M. Rotroff; John Jack; Nathan H Campbell; Scott Clark; Alison A. Motsinger-Reif

BackgroundPGxClean is a new web application that performs quality control analyses for data produced by the Affymetrix DMET chip or other candidate gene technologies. Importantly, the software does not assume that variants are biallelic single-nucleotide polymorphisms, but can be used on the variety of variant characteristics included on the DMET chip. Once quality control analyses has been completed, the associated PGxClean-Viz web application performs principal component analyses and provides tools for characterizing and visualizing population structure.FindingsThe PGxClean web application accepts genotype data from the Affymetrix DMET chip or the PLINK PED format with genotypes annotated as (A,C,G,T or 1,2,3,4). Options for removing missing data and calculating genotype and allele frequencies are offered. Data can be subdivided by cohort characteristics, such as family ID, sex, phenotype, or case–control status. Once the data has been processed through the PGxClean web application, the output files can be entered into the PGxClean-Viz web application for performing principal component analysis to visualize population substructure.ConclusionsThe PGxClean software provides rapid quality-control processing, data analysis, and data visualization for the Affymetrix DMET chip or other candidate gene technologies while improving on common analysis platforms by not assuming that variants are biallelic. The web application is available at http://www.pgxclean.com.

Collaboration


Dive into the John Jack's collaboration.

Top Co-Authors

Avatar

Alison A. Motsinger-Reif

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Howard L. McLeod

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Rotroff

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Tammy M. Havener

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chad Brown

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jon B. Toledo

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge