Tissue specificity-aware TWAS (TSA-TWAS) framework identifies novel associations with metabolic, immunologic, and virologic traits in HIV-positive adults

Binglan Li, Yogasudha Veturi, Anurag Verma, Yuki Bradford, Eric S Daar, Roy M Gulick, Sharon A Riddler, Gregory K Robbins, Jeffrey L Lennox, David W Haas, Marylyn D Ritchie, Binglan Li, Yogasudha Veturi, Anurag Verma, Yuki Bradford, Eric S Daar, Roy M Gulick, Sharon A Riddler, Gregory K Robbins, Jeffrey L Lennox, David W Haas, Marylyn D Ritchie

Abstract

As a type of relatively new methodology, the transcriptome-wide association study (TWAS) has gained interest due to capacity for gene-level association testing. However, the development of TWAS has outpaced statistical evaluation of TWAS gene prioritization performance. Current TWAS methods vary in underlying biological assumptions about tissue specificity of transcriptional regulatory mechanisms. In a previous study from our group, this may have affected whether TWAS methods better identified associations in single tissues versus multiple tissues. We therefore designed simulation analyses to examine how the interplay between particular TWAS methods and tissue specificity of gene expression affects power and type I error rates for gene prioritization. We found that cross-tissue identification of expression quantitative trait loci (eQTLs) improved TWAS power. Single-tissue TWAS (i.e., PrediXcan) had robust power to identify genes expressed in single tissues, but, often found significant associations in the wrong tissues as well (therefore had high false positive rates). Cross-tissue TWAS (i.e., UTMOST) had overall equal or greater power and controlled type I error rates for genes expressed in multiple tissues. Based on these simulation results, we applied a tissue specificity-aware TWAS (TSA-TWAS) analytic framework to look for gene-based associations with pre-treatment laboratory values from AIDS Clinical Trial Group (ACTG) studies. We replicated several proof-of-concept transcriptionally regulated gene-trait associations, including UGT1A1 (encoding bilirubin uridine diphosphate glucuronosyltransferase enzyme) and total bilirubin levels (p = 3.59×10-12), and CETP (cholesteryl ester transfer protein) with high-density lipoprotein cholesterol (p = 4.49×10-12). We also identified several novel genes associated with metabolic and virologic traits, as well as pleiotropic genes that linked plasma viral load, absolute basophil count, and/or triglyceride levels. By highlighting the advantages of different TWAS methods, our simulation study promotes a tissue specificity-aware TWAS analytic framework that revealed novel aspects of HIV-related traits.

Trial registration: ClinicalTrials.gov NCT00000919 NCT00013520 NCT00050895 NCT00118898 NCT00811954.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: G.K.R. has been on principal investigator on research grants to Massachusetts General Hospital from Gilead Sciences, Citius Pharmaceuticals, Emergent Biosolutions, Pfizer and Leonard Meron Bioscience -all research support paid to the institution. E.S.D. has received research grant support from Gilead, Merck and ViiV and has been consultant for Abbvie, Gilead, Merck and Genentech. S.A.R. has been an investigator on research grants to the University of Pittsburgh from Gilead, Merck, and Bristol Myers Squibb and she has been a consultant to Novimab. M.D.R. is on the scientific advisory board for Cipherome and Goldfinch Bio. All other authors have no conflicts to disclose.

Figures

Fig 1. Cross-tissue TWAS simulation scheme.
Fig 1. Cross-tissue TWAS simulation scheme.
With the simulation parameters, we were able to generate SNP-gene-trait relations of varied tissue specificity backgrounds. In each replication, simulated datasets were divided into an eQTL detection dataset and a TWAS dataset. The former was used to identify eQTLs using different eQTL detection methods and the sample size was equivalent to that of GTEx. The detected eQTLs were then passed, separately, to the TWAS dataset to assist gene-level association tests. The TWAS dataset sample size was equivalent of that of the ACTG clinical trial dataset. Two types of gene-level association approaches estimated and ascribed p-values to the simulated gene-trait relations. In each replication, we simulated 100 different SNP-gene-trait pairs for one single point estimation of TWAS gene prioritization performance. All association p-values had been adjusted for the number of genes and tissues in each replication. 20 independent replications were conducted to obtain the distribution of TWAS performance statistics.
Fig 2. Power of different TWAS methods…
Fig 2. Power of different TWAS methods in prioritizing genes of varied tissue specificity properties.
Power was the proportion of successfully identified gene-trait associations in the causal tissue out of all simulations. X-axis is the number of gene-expressing tissues. Each column stands for the proportion of eQTLs that are shared among tissues for a gene. Each row is the similarity of gene expression profiles across tissues which is estimated by correlation. Moving from the top left to the bottom right is a gradient spectrum from tissue-specific genes to broadly expressed genes. The colors represent different TWAS methods and y-axis is the power. For tissue-specific genes at the top left, single-tissue TWAS (Elastic Net-SLR) and cross-tissue TWAS (Group LASSO-GBJ) had similar power. For broadly expressed genes at the bottom right, cross-tissue TWAS (Group LASSO-GBJ) had greater power. Brackets showed pairwise comparison of power between the Group LASSO-GBJ and other TWAS methods using Wilcoxon Signed-rank Test. Black brackets were cases where Group LASSO-GBJ had higher power than other three methods; red brackets were cases where Group LASSO-GBJ had lower power than other three methods. *p-value

Fig 3. Type I error rates of…

Fig 3. Type I error rates of different TWAS methods in prioritizing genes of diverse…

Fig 3. Type I error rates of different TWAS methods in prioritizing genes of diverse tissue specificity properties.
Type I error rate was the probability that TWAS wrongly identified a gene-trait association as significant while there was not any signal simulated in the dataset. Association p-values were controlled for the number of genes and tested tissues. X-axis is the number of gene-expressing tissues. Each column stands for the proportion of eQTLs that are shared among tissues for a gene. Each row is the similarity of gene expression profiles across tissues which is estimated by correlation. Moving from the top left to the bottom right is a gradient spectrum from tissue-specific genes to broadly expressed genes. The colors represent different TWAS methods and y-axis is the type I error rate. All TWAS methods had controlled type I error rates (≤ 5%).

Fig 4. False positive rates of tissues…

Fig 4. False positive rates of tissues among statistically significant results.

False positive rates were…

Fig 4. False positive rates of tissues among statistically significant results.
False positive rates were the proportion of significant associations found in trait-irrelevant tissues amongst all significant results. Association p-values were controlled for the number of genes and tested tissues. X-axis is the number of gene-expressing tissues. Each column stands for the proportion of eQTLs that are shared among tissues for a gene. Each row is the similarity of gene expression profiles across tissues which is estimated by correlation. Moving from the top left to the bottom right is a gradient spectrum from tissue-specific genes to broadly expressed genes. Colors represent different TWAS methods and y-axis is the false positive rate of tissues among statistically significant results. Single-tissue TWAS wrongly identified 5% and 77% trait-irrelevant tissues for tissue-specific genes and broadly expressed genes, respectively.

Fig 5. A proposed TSA-TWAS analytic framework…

Fig 5. A proposed TSA-TWAS analytic framework that leverages TWAS performance on genes of different…

Fig 5. A proposed TSA-TWAS analytic framework that leverages TWAS performance on genes of different tissue specificity properties.
The framework proposed based on our simulations is as follows: If trait-related tissue(s) are known for a trait or disease of interest, run single-tissue TWAS, for example, PrediXcan. If trait-related tissue(s) are unknown, run cross-tissue TWAS (UTMOST) on the genes that are expressed in more than one tissue and run single-tissue TWAS (PrediXcan) on the genes that are expressed in one single tissue.

Fig 6. Power of the TSA-TWAS framework…

Fig 6. Power of the TSA-TWAS framework when there were different proportions of tissue-specific genes…

Fig 6. Power of the TSA-TWAS framework when there were different proportions of tissue-specific genes in the data.
The power of TSA-TWAS was compared to only running single-tissue TWAS (elastic net-SLR) and cross-tissue TWAS (Group LASSO-GBJ test). TSA-TWAS had consistent power of identifying complex trait-related genes and was robust to makeups of tissue-specific and multi-tissue genes in a dataset.

Fig 7. The TSA-TWAS analytic framework for…

Fig 7. The TSA-TWAS analytic framework for the ACTG combined genotyping phase I-IV baseline laboratory…

Fig 7. The TSA-TWAS analytic framework for the ACTG combined genotyping phase I-IV baseline laboratory traits.
Approximately 2.2 million SNPs, 4,360 individuals, and 37 baseline laboratory traits survived the QC. UTMOST eQTL models were used to impute GReX of a total of 12,038 genes in 49 tissues. 2,812 genes (23%) had GReX in one single tissue, and 9,226 genes (77%) had GReX in more than one tissue.

Fig 8. PhenoGram of statistically significant gene-trait…

Fig 8. PhenoGram of statistically significant gene-trait associations identified by the TSA-TWAS analytic framework.

We…

Fig 8. PhenoGram of statistically significant gene-trait associations identified by the TSA-TWAS analytic framework.
We plotted the associations with p-value −7. Each association is arranged according to the SNP location on each chromosome and the points are color-coded by baseline laboratory values. Diamonds represented previously reported or replicated associations, and circle represented novel associations identified in this study.
All figures (8)
Fig 3. Type I error rates of…
Fig 3. Type I error rates of different TWAS methods in prioritizing genes of diverse tissue specificity properties.
Type I error rate was the probability that TWAS wrongly identified a gene-trait association as significant while there was not any signal simulated in the dataset. Association p-values were controlled for the number of genes and tested tissues. X-axis is the number of gene-expressing tissues. Each column stands for the proportion of eQTLs that are shared among tissues for a gene. Each row is the similarity of gene expression profiles across tissues which is estimated by correlation. Moving from the top left to the bottom right is a gradient spectrum from tissue-specific genes to broadly expressed genes. The colors represent different TWAS methods and y-axis is the type I error rate. All TWAS methods had controlled type I error rates (≤ 5%).
Fig 4. False positive rates of tissues…
Fig 4. False positive rates of tissues among statistically significant results.
False positive rates were the proportion of significant associations found in trait-irrelevant tissues amongst all significant results. Association p-values were controlled for the number of genes and tested tissues. X-axis is the number of gene-expressing tissues. Each column stands for the proportion of eQTLs that are shared among tissues for a gene. Each row is the similarity of gene expression profiles across tissues which is estimated by correlation. Moving from the top left to the bottom right is a gradient spectrum from tissue-specific genes to broadly expressed genes. Colors represent different TWAS methods and y-axis is the false positive rate of tissues among statistically significant results. Single-tissue TWAS wrongly identified 5% and 77% trait-irrelevant tissues for tissue-specific genes and broadly expressed genes, respectively.
Fig 5. A proposed TSA-TWAS analytic framework…
Fig 5. A proposed TSA-TWAS analytic framework that leverages TWAS performance on genes of different tissue specificity properties.
The framework proposed based on our simulations is as follows: If trait-related tissue(s) are known for a trait or disease of interest, run single-tissue TWAS, for example, PrediXcan. If trait-related tissue(s) are unknown, run cross-tissue TWAS (UTMOST) on the genes that are expressed in more than one tissue and run single-tissue TWAS (PrediXcan) on the genes that are expressed in one single tissue.
Fig 6. Power of the TSA-TWAS framework…
Fig 6. Power of the TSA-TWAS framework when there were different proportions of tissue-specific genes in the data.
The power of TSA-TWAS was compared to only running single-tissue TWAS (elastic net-SLR) and cross-tissue TWAS (Group LASSO-GBJ test). TSA-TWAS had consistent power of identifying complex trait-related genes and was robust to makeups of tissue-specific and multi-tissue genes in a dataset.
Fig 7. The TSA-TWAS analytic framework for…
Fig 7. The TSA-TWAS analytic framework for the ACTG combined genotyping phase I-IV baseline laboratory traits.
Approximately 2.2 million SNPs, 4,360 individuals, and 37 baseline laboratory traits survived the QC. UTMOST eQTL models were used to impute GReX of a total of 12,038 genes in 49 tissues. 2,812 genes (23%) had GReX in one single tissue, and 9,226 genes (77%) had GReX in more than one tissue.
Fig 8. PhenoGram of statistically significant gene-trait…
Fig 8. PhenoGram of statistically significant gene-trait associations identified by the TSA-TWAS analytic framework.
We plotted the associations with p-value −7. Each association is arranged according to the SNP location on each chromosome and the points are color-coded by baseline laboratory values. Diamonds represented previously reported or replicated associations, and circle represented novel associations identified in this study.

References

    1. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, et al.. 10 Years of GWAS Discovery: Biology, Function, and Translation. The American Journal of Human Genetics. ElsevierCompany; 2017. July 6;101(1):5–22. 10.1016/j.ajhg.2017.06.005
    1. Lappalainen T. Functional genomics bridges the gap between quantitative genetics and molecular biology. Genome Research. Cold Spring Harbor Lab; 2015. October;25(10):1427–31. 10.1101/gr.190983.115
    1. MacArthur J, Bowler E, Cerezo M, Gil L, Hall P, Hastings E, et al.. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Research. 2017. January 4;45(D1):D896–D901. 10.1093/nar/gkw1133
    1. Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, et al.. Systematic localization of common disease-associated variation in regulatory DNA. Science. American Association for the Advancement of Science; 2012. September 7;337(6099):1190–5. 10.1126/science.1222794
    1. Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, et al.. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet. Nature Publishing Group; 2015. August 10;47(9):1091–8. 10.1038/ng.3367
    1. Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, et al.. Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet. Nature Publishing Group; 2016. March;48(3):245–52. 10.1038/ng.3506
    1. Thériault S, Gaudreault N, Lamontagne M, Rosa M, Boulanger M-C, Messika-Zeitoun D, et al.. A transcriptome-wide association study identifies PALMD as a susceptibility gene for calcific aortic valve stenosis. Nature Communications. Nature Publishing Group; 2018. March 7;9(1):988. 10.1038/s41467-018-03260-6
    1. Wu L, Shi W, Long J, Guo X, Michailidou K, Beesley J, et al.. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer. Nat Genet. Nature Publishing Group; 2018. June 18;50(7):968–78. 10.1038/s41588-018-0132-x
    1. Mancuso N, Shi H, Goddard P, Kichaev G, Gusev A, Pasaniuc B. Integrating Gene Expression with Summary Association Statistics to Identify Genes Associated with 30 Complex Traits. American journal of human genetics. Elsevier; 2017. March 2;100(3):473–87. 10.1016/j.ajhg.2017.01.031
    1. Battle A, Brown CD, Engelhardt BE, Montgomery SB. Genetic effects on gene expression across human tissues. Nature. Nature Publishing Group; 2017. October 12;550(7675):204–13. 10.1038/nature24277
    1. Võsa U, Claringbould A, Westra H-J, Bonder MJ, Deelen P, Zeng B, et al.. Unraveling the polygenic architecture of complex traits using blood eQTL meta-analysis. 2018.
    1. Li B, Veturi Y, Bradford Y, Verma SS, Verma A, Lucas AM, et al.. Influence of tissue context on gene prioritization for predicted transcriptome-wide association studies. Pac Symp Biocomput. 2019;24:296–307.
    1. Flutre T, Wen X, Pritchard J, Stephens M. A statistical framework for joint eQTL analysis in multiple tissues. Gibson G, editor. PLoS Genet. Public Library of Science; 2013. May;9(5):e1003486. 10.1371/journal.pgen.1003486
    1. Sul JH, Han B, Ye C, Choi T, Eskin E. Effectively identifying eQTLs from multiple tissues by combining mixed model and meta-analytic approaches. Schork NJ, editor. PLoS Genet. Public Library of Science; 2013. June;9(6):e1003491. 10.1371/journal.pgen.1003491
    1. Hu Y, Li M, Lu Q, Weng H, Wang J, Zekavat SM, et al.. A statistical framework for cross-tissue transcriptome-wide association analysis. Nat Genet. Nature Publishing Group; 2019. March;51(3):568–76. 10.1038/s41588-019-0345-7
    1. Barbeira AN, Pividori M, Zheng J, Wheeler HE, Nicolae DL, Im HK. Integrating predicted transcriptome from multiple tissues improves association detection. Plagnol V, editor. PLoS Genet. 2019. January;15(1):e1007889. 10.1371/journal.pgen.1007889
    1. Liu X, Finucane HK, Gusev A, Bhatia G, Gazal S, O’Connor L, et al.. Functional Architectures of Local and Distal Regulation of Gene Expression in Multiple Human Tissues. American journal of human genetics. 2017. April 6;100(4):605–16. 10.1016/j.ajhg.2017.03.002
    1. Wainberg M, Sinnott-Armstrong N, Mancuso N, Barbeira AN, Knowles DA, Golan D, et al.. Opportunities and challenges for transcriptome-wide association studies. Nat Genet. Nature Publishing Group; 2019. April;51(4):592–9. 10.1038/s41588-019-0385-z
    1. Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, et al.. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. American Society for Biochemistry and Molecular Biology; 2014. February;13(2):397–406.
    1. Kryuchkova-Mostacci N, Robinson-Rechavi M. Tissue-Specificity of Gene Expression Diverges Slowly between Orthologs, and Rapidly between Paralogs. Ouzounis CA, editor. PLoS Comput Biol. Public Library of Science; 2016. December;12(12):e1005274. 10.1371/journal.pcbi.1005274
    1. Veturi Y, Ritchie MD. How powerful are summary-based methods for identifying expression-trait associations under different genetic architectures? Pac Symp Biocomput. 2018;23:228–39.
    1. Wheeler HE, Shah KP, Brenner J, Garcia T, Aquino-Michaels K, GTEx Consortium, et al.. Survey of the Heritability and Sparse Architecture of Gene Expression Traits across Human Tissues. Montgomery SB, editor. PLoS Genet. 2016. November 11;12(11):e1006423–3. 10.1371/journal.pgen.1006423
    1. Ongen H, Brown AA, Delaneau O, Panousis NI, Nica AC, GTEx Consortium, et al.. Estimating the causal tissues for complex traits and diseases. Nat Genet. 2017. December;49(12):1676–83. 10.1038/ng.3981
    1. Mancuso N, Freund MK, Johnson R, Shi H, Kichaev G, Gusev A, et al.. Probabilistic fine-mapping of transcriptome-wide association studies. Nat Genet. 2019. April;51(4):675–82. 10.1038/s41588-019-0367-1
    1. Moore CB, Verma A, Pendergrass S, Verma SS, Johnson DH, Daar ES, et al.. Phenome-wide Association Study Relating Pretreatment Laboratory Parameters With Human Genetic Variants in AIDS Clinical Trials Group Protocols. Open Forum Infect Dis. 2015. January;2(1):ofu113. 10.1093/ofid/ofu113
    1. Verma A, Bradford Y, Verma SS, Pendergrass SA, Daar ES, Venuto C, et al.. Multiphenotype association study of patients randomized to initiate antiretroviral regimens in AIDS Clinical Trials Group protocol A5202. Pharmacogenetics and Genomics. 2017. March;27(3):101–11. 10.1097/FPC.0000000000000263
    1. Coltell O, Asensio EM, Sorlí JV, Barragán R, Fernández-Carrión R, Portolés O, et al.. Genome-Wide Association Study (GWAS) on Bilirubin Concentrations in Subjects with Metabolic Syndrome: Sex-Specific GWAS Analysis and Gene-Diet Interactions in a Mediterranean Population. Nutrients. Multidisciplinary Digital Publishing Institute; 2019. January 4;11(1):90.
    1. Dai X, Wu C, He Y, Gui L, Zhou L, Guo H, et al.. A genome-wide association study for serum bilirubin levels and gene-environment interaction in a Chinese population. Genet Epidemiol. 2013. April;37(3):293–300. 10.1002/gepi.21711
    1. Tukey RH, Strassburg CP. Human UDP-glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol. 2000;40:581–616. 10.1146/annurev.pharmtox.40.1.581
    1. Barter PJ, H Bryan Brewer J, Chapman MJ, Hennekens CH, Rader DJ, Tall AR. Cholesteryl Ester Transfer Protein. Arterioscler Thromb Vasc Biol. Lippincott Williams & Wilkins; 2003. February 1;23(2):160–7. 10.1161/01.atv.0000054658.91146.64
    1. Chambers JC, Zhang W, Sehmi J, Li X, Wass MN, Van der Harst P, et al.. Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma. Nat Genet. 2011. October 16;43(11):1131–8. 10.1038/ng.970
    1. Kanai M, Akiyama M, Takahashi A, Matoba N, Momozawa Y, Ikeda M, et al.. Genetic analysis of quantitative traits in the Japanese population links cell types to complex human diseases. Nat Genet. Nature Publishing Group; 2018. March;50(3):390–400. 10.1038/s41588-018-0047-6
    1. Le Clerc S, Coulonges C, Delaneau O, van Manen D, Herbeck JT, Limou S, et al.. Screening low-frequency SNPS from genome-wide association study reveals a new risk allele for progression to AIDS. J Acquir Immune Defic Syndr. 2011. March 1;56(3):279–84. 10.1097/QAI.0b013e318204982b
    1. Rhee EP, Ho JE, Chen M-H, Shen D, Cheng S, Larson MG, et al.. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013. July 2;18(1):130–43. 10.1016/j.cmet.2013.06.013
    1. Lingwood CA, Branch DR. The role of glycosphingolipids in HIV/AIDS. Discov Med. Discov Med; 2011. April;11(59):303–13.
    1. van Til NP, Heutinck KM, van der Rijt R, Paulusma CC, van Wijland M, Markusic DM, et al.. Alteration of viral lipid composition by expression of the phospholipid floppase ABCB4 reduces HIV vector infectivity. Retrovirology. BioMed Central; 2008. February 1;5(1):14–9. 10.1186/1742-4690-5-14
    1. Wu B, Ouyang Z, Lyon CJ, Zhang W, Clift T, Bone CR, et al.. Plasma Levels of Complement Factor I and C4b Peptides Are Associated with HIV Suppression. ACS Infect Dis. 2017. December 8;3(12):880–5. 10.1021/acsinfecdis.7b00042
    1. Dunn SJ, Khan IH, Chan UA, Scearce RL, Melara CL, Paul AM, et al.. Identification of cell surface targets for HIV-1 therapeutics using genetic screens. Virology. 2004. April 10;321(2):260–73. 10.1016/j.virol.2004.01.010
    1. Migueles SA, Sabbaghian MS, Shupert WL, Bettinotti MP, Marincola FM, Martino L, et al.. HLA B*5701 is highly associated with restriction of virus replication in a subgroup of HIV-infected long term nonprogressors. PNAS. 2000. March 14;97(6):2709–14. 10.1073/pnas.050567397
    1. Kjolby M, Andersen OM, Breiderhoff T, Fjorback AW, Pedersen KM, Madsen P, et al.. Sort1, encoded by the cardiovascular risk locus 1p13.3, is a regulator of hepatic lipoprotein export. Cell Metab. 2010. September 8;12(3):213–23. 10.1016/j.cmet.2010.08.006
    1. Arvind P, Nair J, Jambunathan S, Kakkar VV, Shanker J. CELSR2-PSRC1-SORT1 gene expression and association with coronary artery disease and plasma lipid levels in an Asian Indian cohort. J Cardiol. 2014. November;64(5):339–46. 10.1016/j.jjcc.2014.02.012
    1. Musunuru K, Strong A, Frank-Kamenetsky M, Lee NE, Ahfeldt T, Sachs KV, et al.. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature Publishing Group. Nature Publishing Group; 2010. August 5;466(7307):714–9. 10.1038/nature09266
    1. Akay C, Lindl KA, Shyam N, Nabet B, Goenaga-Vazquez Y, Ruzbarsky J, et al.. Activation status of integrated stress response pathways in neurones and astrocytes of HIV-associated neurocognitive disorders (HAND) cortex. Neuropathol Appl Neurobiol. John Wiley & Sons, Ltd; 2012. April;38(2):175–200. 10.1111/j.1365-2990.2011.01215.x
    1. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992. May;74(5):1045–52. 10.1210/jcem.74.5.1373735
    1. Michalak P, Coexpression, coregulation, and cofunctionality of neighboring genes in eukaryotic genomes. Genomics. 2008. March;91(3):243–8. 10.1016/j.ygeno.2007.11.002
    1. Pividori M, Rajagopal PS, Barbeira A, Liang Y, Melia O, Bastarache L, et al.. PhenomeXcan: Mapping the genome to the phenome through the transcriptome. Sci Adv. 2020. September;6(37). 10.1126/sciadv.aba2083
    1. Barbeira AN, Bonazzola R, Gamazon ER, Liang Y, Park Y, Kim-Hellmuth S, et al.. Exploiting the GTEx resources to decipher the mechanisms at GWAS loci. bioRxiv. Cold Spring Harbor Laboratory; 2020. May 23;42(D1):814350.
    1. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. American journal of human genetics. 2011. January 7;88(1):76–82. 10.1016/j.ajhg.2010.11.011
    1. Robbins GK, De Gruttola V, Shafer RW, Smeaton LM, Snyder SW, Pettinelli C, et al.. Comparison of sequential three-drug regimens as initial therapy for HIV-1 infection. N Engl J Med. Massachusetts Medical Society; 2003. December 11;349(24):2293–303. 10.1056/NEJMoa030264
    1. Gulick RM, Ribaudo HJ, Shikuma CM, Lustgarten S, Squires KE, Meyer WA, et al.. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N Engl J Med. Massachusetts Medical Society; 2004. April 29;350(18):1850–61. 10.1056/NEJMoa031772
    1. Gulick RM, Ribaudo HJ, Shikuma CM, Lalama C, Schackman BR, Meyer WA, et al.. Three- vs four-drug antiretroviral regimens for the initial treatment of HIV-1 infection: a randomized controlled trial. JAMA. 2006. August 16;296(7):769–81. 10.1001/jama.296.7.769
    1. Riddler SA, Haubrich R, DiRienzo AG, Peeples L, Powderly WG, Klingman KL, et al.. Class-sparing regimens for initial treatment of HIV-1 infection. N Engl J Med. Massachusetts Medical Society; 2008. May 15;358(20):2095–106. 10.1056/NEJMoa074609
    1. Sax PE, Tierney C, Collier AC, Fischl MA, Mollan K, Peeples L, et al.. Abacavir-lamivudine versus tenofovir-emtricitabine for initial HIV-1 therapy. N Engl J Med. Massachusetts Medical Society; 2009. December 3;361(23):2230–40. 10.1056/NEJMoa0906768
    1. Daar ES, Tierney C, Fischl MA, Sax PE, Mollan K, Budhathoki C, et al.. Atazanavir Plus Ritonavir or Efavirenz as Part of a 3-Drug Regimen for Initial Treatment of HIV-1: A Randomized Trial. Ann Intern Med. American College of Physicians; 2011. April 5;154(7):445–56. 10.7326/0003-4819-154-7-201104050-00316
    1. Lennox JL, Landovitz RJ, Ribaudo HJ, Ofotokun I, Na LH, Godfrey C, et al.. A Phase III Comparative Study of the Efficacy and Tolerability of Three Non-Nucleoside Reverse Transcriptase Inhibitor-Sparing Antiretroviral Regimens for Treatment-Naïve HIV-1-Infected Volunteers: A Randomized, Controlled Trial. Ann Intern Med. NIH Public Access; 2014. October 7;161(7):461–71. 10.7326/M14-1084
    1. Turner S, Armstrong LL, Bradford Y, Carlson CS, Crawford DC, Crenshaw AT, et al.. Quality control procedures for genome-wide association studies. Haines JL, Korf BR, Morton CC, Seidman CE, Seidman JG, Smith DR, editors. Curr Protoc Hum Genet. 2011. January;Chapter 1(1):Unit1.19–1.19.18. 10.1002/0471142905.hg0119s68
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, et al.. PLINK: a tool set for whole-genome association and population-based linkage analyses. The American Journal of Human Genetics. 2007. September;81(3):559–75. 10.1086/519795
    1. Howie BN, Donnelly P, Marchini J. A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies. Schork NJ, editor. PLoS Genet. Public Library of Science; 2009. June 19;5(6):e1000529. 10.1371/journal.pgen.1000529
    1. 1000 Genomes Project Consortium, Abecasis GR, Altshuler D, Auton A, Brooks LD, Durbin RM, et al. A map of human genome variation from population-scale sequencing. Nature Publishing Group. 2010. October 28;467(7319):1061–73. 10.1038/nature09534
    1. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. Nature Publishing Group; 2006. August;38(8):904–9. 10.1038/ng1847
    1. Lucas AM, Palmiero NE, McGuigan J, Passero K, Zhou J, Orie D, et al.. CLARITE Facilitates the Quality Control and Analysis Process for EWAS of Metabolic-Related Traits. Front Genet. Frontiers; 2019. December 18;10:1164. 10.3389/fgene.2019.01164
    1. Langsted A, Nordestgaard BG. Nonfasting versus fasting lipid profile for cardiovascular risk prediction. Pathology. 2019. February;51(2):131–41. 10.1016/j.pathol.2018.09.062
    1. Nordestgaard BG. A Test in Context: Lipid Profile, Fasting Versus Nonfasting. J Am Coll Cardiol. 2017. September 26;70(13):1637–46. 10.1016/j.jacc.2017.08.006
    1. Mora S, Chang CL, Moorthy MV, Sever PS. Association of Nonfasting vs Fasting Lipid Levels With Risk of Major Coronary Events in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid Lowering Arm. JAMA Intern Med. American Medical Association; 2019. May 28;179(7):898–905. 10.1001/jamainternmed.2019.0392
    1. Barbeira AN, Bonazzola R, Gamazon ER, Liang Y, Park Y, Kim-Hellmuth S, et al.. Exploiting the GTEx resources to decipher the mechanisms at GWAS loci. bioRxiv. Cold Spring Harbor Laboratory; 2020. May 23;42(D1):814350.
    1. Hall MA, Wallace J, Lucas A, Kim D, Basile AO, Verma SS, et al.. PLATO software provides analytic framework for investigating complexity beyond genome-wide association studies. Nature Communications. Nature Publishing Group; 2017. October 27;8(1):1167. 10.1038/s41467-017-00802-2
    1. Grady BJ, Torstenson E, Dudek SM, Giles J, Sexton D, Ritchie MD. Finding unique filter sets in PLATO: a precursor to efficient interaction analysis in GWAS data. Pac Symp Biocomput. 2010:315–26.
    1. Wolfe D, Dudek S, Ritchie MD, Pendergrass SA. Visualizing genomic information across chromosomes with PhenoGram. BioData Min. BioMed Central; 2013. October 16;6(1):18–12. 10.1186/1756-0381-6-18
    1. Wen X. Effective QTL Discovery Incorporating Genomic Annotations. bioRxiv. Cold Spring Harbor Laboratory; 2015. November 16:032003.

Source: PubMed

3
구독하다