85 results on '"Andrew D. Skol"'
Search Results
2. Gene-Expression-Guided Selection of Candidate Loci and Molecular Phenotype Analyses Enhance Genetic Discovery in Systemic Lupus Erythematosus
- Author
-
Yelena Koldobskaya, Kichul Ko, Akaash A. Kumar, Sandra Agik, Jasmine Arrington, Silvia N. Kariuki, Beverly S. Franek, Marissa Kumabe, Tammy O. Utset, Meenakshi Jolly, Andrew D. Skol, and Timothy B. Niewold
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disorder characterized by differences in autoantibody profiles, serum cytokines, and clinical manifestations. We have previously conducted a case-case genome-wide association study (GWAS) of SLE patients to detect associations with autoantibody profile and serum interferon alpha (IFN-α). In this study, we used public gene expression data sets to rationally select additional single nucleotide polymorphisms (SNPs) for validation. The top 200 GWAS SNPs were searched in a database which compares genome-wide expression data to genome-wide SNP genotype data in HapMap cell lines. SNPs were chosen for validation if they were associated with differential expression of 15 or more genes at a significance of P
- Published
- 2012
- Full Text
- View/download PDF
3. Integration of genomics and transcriptomics predicts diabetic retinopathy susceptibility genes
- Author
-
Andrew D Skol, Segun C Jung, Ana Marija Sokovic, Siquan Chen, Sarah Fazal, Olukayode Sosina, Poulami P Borkar, Amy Lin, Maria Sverdlov, Dingcai Cao, Anand Swaroop, Ionut Bebu, DCCT/EDIC Study group, Barbara E Stranger, and Michael A Grassi
- Subjects
LCLs ,folliculin ,diabetic retinopathy ,eQTL ,mendelian randomization ,gene expression ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
We determined differential gene expression in response to high glucose in lymphoblastoid cell lines derived from matched individuals with type 1 diabetes with and without retinopathy. Those genes exhibiting the largest difference in glucose response were assessed for association with diabetic retinopathy in a genome-wide association study meta-analysis. Expression quantitative trait loci (eQTLs) of the glucose response genes were tested for association with diabetic retinopathy. We detected an enrichment of the eQTLs from the glucose response genes among small association p-values and identified folliculin (FLCN) as a susceptibility gene for diabetic retinopathy. Expression of FLCN in response to glucose was greater in individuals with diabetic retinopathy. Independent cohorts of individuals with diabetes revealed an association of FLCN eQTLs with diabetic retinopathy. Mendelian randomization confirmed a direct positive effect of increased FLCN expression on retinopathy. Integrating genetic association with gene expression implicated FLCN as a disease gene for diabetic retinopathy.
- Published
- 2020
- Full Text
- View/download PDF
4. Supplementary Tables from Functional Common and Rare ERBB2 Germline Variants Cooperate in Familial and Sporadic Cancer Susceptibility
- Author
-
Kenan Onel, Jaume Mora, Zeynep H. Gümüş, Robert J. Klein, Hector Salvador, Cinzia Lavarino, Andrew D. Skol, Lei Huang, Hyesan Lee, Alyna Katti, Myvizhi Esai Selvan, Mark Sasaki, Anita Ng, and Riyue Bao
- Abstract
Supplementary Tables 1-7
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 1 from Functional Common and Rare ERBB2 Germline Variants Cooperate in Familial and Sporadic Cancer Susceptibility
- Author
-
Kenan Onel, Jaume Mora, Zeynep H. Gümüş, Robert J. Klein, Hector Salvador, Cinzia Lavarino, Andrew D. Skol, Lei Huang, Hyesan Lee, Alyna Katti, Myvizhi Esai Selvan, Mark Sasaki, Anita Ng, and Riyue Bao
- Abstract
Figure S1
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Functional Common and Rare ERBB2 Germline Variants Cooperate in Familial and Sporadic Cancer Susceptibility
- Author
-
Kenan Onel, Jaume Mora, Zeynep H. Gümüş, Robert J. Klein, Hector Salvador, Cinzia Lavarino, Andrew D. Skol, Lei Huang, Hyesan Lee, Alyna Katti, Myvizhi Esai Selvan, Mark Sasaki, Anita Ng, and Riyue Bao
- Abstract
We investigated a Spanish and Catalan family in which multiple cancer types tracked across three generations, but for which no genetic etiology had been identified. Whole-exome sequencing of germline DNA from multiple affected family members was performed to identify candidate variants to explain this occurrence of familial cancer. We discovered in all cancer-affected family members a single rare heterozygous germline variant (I654V, rs1801201) in ERBB2/HER2, which is located in a transmembrane glycine zipper motif critical for ERBB2-mediated signaling and in complete linkage disequilibrium (D' = 1) with a common polymorphism (I655V, rs1136201) previously reported in some populations as associated with cancer risk. Because multiple cancer types occurred in this family, we tested both the I654V and the I655V variants for association with cancer across multiple tumor types in 6,371 cases of Northern European ancestry drawn from The Cancer Genome Atlas and 6,647 controls, and found that the rare variant (I654V) was significantly associated with an increased risk for cancer (OR = 1.40; P = 0.021; 95% confidence interval (CI), 1.05–1.89). Functional assays performed in HEK 293T cells revealed that both the I655V single mutant (SM) and the I654V;I655V double mutant (DM) stabilized ERBB2 protein and activated ERBB2 signaling, with the DM activating ERBB2 significantly more than the SM alone. Thus, our results suggest a model whereby heritable genetic variation in the transmembrane domain activating ERBB2 signaling is associated with both sporadic and familial cancer risk, with increased ERBB2 stabilization and activation associated with increased cancer risk.Prevention Relevance:By performing whole-exome sequencing on germline DNA from multiple cancer-affected individuals belonging to a family in which multiple cancer types track across three generations, we identified and then characterized functional common and rare variation in ERBB2 associated with both sporadic and familial cancer. Our results suggest that heritable variation activating ERBB2 signaling is associated with risk for multiple cancer types, with increases in signaling correlated with increases in risk, and modified by ancestry or family history.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib
- Author
-
Federico Innocenti, Kari K. Alitalo, Nancy Klauber-DeMore, Yuri K. Peterson, Carol E. Peña, Eleanor Hilliard, Amy S. Etheridge, Dylan M. Glubb, Veli-Matti Leppänen, Andrew D. Skol, and Daniel J. Crona
- Abstract
Molecular markers of sorafenib efficacy in patients with metastatic renal cell carcinoma (mRCC) are not available. The purpose of this study was to discover genetic markers of survival in patients with mRCC treated with sorafenib. Germline variants from 56 genes were genotyped in 295 patients with mRCC. Variant-overall survival (OS) associations were tested in multivariate regression models. Mechanistic studies were conducted to validate clinical associations. VEGFA rs1885657, ITGAV rs3816375, and WWOX rs8047917 (sorafenib arm), and FLT4 rs307826 and VEGFA rs3024987 (sorafenib and placebo arms combined) were associated with shorter OS. FLT4 rs307826 increased VEGFR-3 phosphorylation, membrane trafficking, and receptor activation. VEGFA rs1885657 and rs58159269 increased transcriptional activity of the constructs containing these variants in endothelial and RCC cell lines, and VEGFA rs58159269 increased endothelial cell proliferation and tube formation. FLT4 rs307826 and VEGFA rs58159269 led to reduced sorafenib cytotoxicity. Genetic variation in VEGFA and FLT4 could affect survival in sorafenib-treated patients with mRCC. These markers should be examined in additional malignancies treated with sorafenib and in other angiogenesis inhibitors used in mRCC.Significance:Clinical and mechanistic data identify germline genetic variants in VEGFA and FLT4 as markers of survival in patients with metastatic renal cell carcinoma.
- Published
- 2023
- Full Text
- View/download PDF
8. Table S1 from Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib
- Author
-
Federico Innocenti, Kari K. Alitalo, Nancy Klauber-DeMore, Yuri K. Peterson, Carol E. Peña, Eleanor Hilliard, Amy S. Etheridge, Dylan M. Glubb, Veli-Matti Leppänen, Andrew D. Skol, and Daniel J. Crona
- Abstract
Genotyped genes.
- Published
- 2023
- Full Text
- View/download PDF
9. Figure S3 from Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib
- Author
-
Federico Innocenti, Kari K. Alitalo, Nancy Klauber-DeMore, Yuri K. Peterson, Carol E. Peña, Eleanor Hilliard, Amy S. Etheridge, Dylan M. Glubb, Veli-Matti Leppänen, Andrew D. Skol, and Daniel J. Crona
- Abstract
Expression of WT and T494A VEGFR-3 in HUVECs (A) and replication of phosphorylation experiments testing the effect of FLT4 rs307826 (A>G, T494A) on VEGFR-3 phosphorylation (B).
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Tables 1-6 from Integrated Genomic Analysis Suggests MLL3 Is a Novel Candidate Susceptibility Gene for Familial Nasopharyngeal Carcinoma
- Author
-
Kenan Onel, Ezra E.W. Cohen, Everett E. Vokes, Rachelle Chambers, Lindsay V. Rhodes, Riyue Bao, Andrew D. Skol, and Mark M. Sasaki
- Abstract
Supplementary Tables 1-6. Supplementary Table 1. List of genes and variants previously reported as associated with NPC. Supplementary Table 2. Proportion of exome covered at different read depths for each sample sequenced. Supplementary Table 3. Variant subtype for each samples sequenced. Supplementary Table 4. Rare deleterious variants shared in the germline of all three NPC-affected family members. Supplementary Table 6. EBV insertion events identified in the exome of each NPC-affected family member.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Methods and Materials from Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib
- Author
-
Federico Innocenti, Kari K. Alitalo, Nancy Klauber-DeMore, Yuri K. Peterson, Carol E. Peña, Eleanor Hilliard, Amy S. Etheridge, Dylan M. Glubb, Veli-Matti Leppänen, Andrew D. Skol, and Daniel J. Crona
- Abstract
SNP selection criteria, imputation and bioinformatic analyses
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 5 from Integrated Genomic Analysis Suggests MLL3 Is a Novel Candidate Susceptibility Gene for Familial Nasopharyngeal Carcinoma
- Author
-
Kenan Onel, Ezra E.W. Cohen, Everett E. Vokes, Rachelle Chambers, Lindsay V. Rhodes, Riyue Bao, Andrew D. Skol, and Mark M. Sasaki
- Abstract
Supplementary Table 5: Somatic Mutations Identified in Individual I-1.
- Published
- 2023
- Full Text
- View/download PDF
13. PIK3R5 genetic predictors of hypertension induced by VEGF-pathway inhibitors
- Author
-
Stefanie Denning, Andrew D. Skol, Alessandro Racioppi, Federico Innocenti, Julia C F Quintanilha, Daniel J. Crona, Jin Wang, Carol Peña, Danyu Lin, and Amy S. Etheridge
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Oncology ,medicine.medical_specialty ,Genome-wide association study ,Single-nucleotide polymorphism ,Significant snps ,Polymorphism, Single Nucleotide ,Risk Assessment ,Linkage Disequilibrium ,Article ,Double-Blind Method ,Predictive Value of Tests ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Testing ,Aged ,Predictive biomarker ,Pharmacology ,Predictive marker ,business.industry ,Middle Aged ,Bevacizumab ,Clinical trial ,Increased risk ,Vegf pathway ,Hypertension ,Molecular Medicine ,Female ,Phosphatidylinositol 3-Kinase ,business ,Biomarkers ,Signal Transduction - Abstract
No biomarkers are available to predict patients at risk of developing hypertension induced by VEGF-pathway inhibitors. This study aimed to identify predictive biomarkers of hypertension induced by these drugs using a discovery-replication approach. The discovery set included 140 sorafenib-treated patients (TARGET study) genotyped for 973 SNPs in 56 genes. The most statistically significant SNPs associated with grade ≥2 hypertension were tested for association with grade ≥2 hypertension in the replication set of a GWAS of 1039 bevacizumab-treated patients from four clinical trials (CALGB/Alliance). In the discovery set, rs444904 (G > A) in PIK3R5 was associated with an increased risk of sorafenib-induced hypertension (p = 0.006, OR = 3.88 95% CI 1.54–9.81). In the replication set, rs427554 (G > A) in PIK3R5 (in complete linkage disequilibrium with rs444904) was associated with an increased risk of bevacizumab-induced hypertension (p = 0.008, OR = 1.39, 95% CI 1.09–1.78). This study identified a predictive marker of drug-induced hypertension that should be evaluated for other VEGF-pathway inhibitors. ClinicalTrials.gov Identifier:NCT00073307 (TARGET).
- Published
- 2021
- Full Text
- View/download PDF
14. Inherited cancer predisposing mutations in patients with therapy-related myeloid neoplasms
- Author
-
Andrew J. Shih, Tomi Jun, Andrew D. Skol, Riyue Bao, Lei Huang, Sapana Vora, Megan E. McNerney, Eric A. Hungate, Michelle M. Le Beau, Richard A. Larson, Aaron Elliott, Hsiao‐Mei Lu, Robert Huether, Felicia Hernandez, Friedrich Stölzel, James M. Allan, and Kenan Onel
- Subjects
Hematology - Abstract
Some patients with therapy-related myeloid neoplasms (t-MN) may have unsuspected inherited cancer predisposition syndrome (CPS). We propose a set of clinical criteria to identify t-MN patients with high risk of CPS (HR-CPS). Among 225 t-MN patients with an antecedent non-myeloid malignancy, our clinical criteria identified 52 (23%) HR-CPS patients. Germline whole-exome sequencing identified pathogenic or likely pathogenic variants in 10 of 27 HR-CPS patients compared to 0 of 9 low-risk CPS patients (37% vs. 0%, p = 0.04). These simple clinical criteria identify t-MN patients most likely to benefit from genetic testing for inherited CPS.
- Published
- 2022
15. Genome-wide association study identifies susceptibility loci for acute myeloid leukemia
- Author
-
Anne M. Dickinson, Gail Jones, David C. Linch, Clare Lendrem, David Grimwade, Richard A. Larson, Andrew D. Skol, Yaobo Xu, Adam Ivey, Wei-Yu Lin, Manja Meggendorfer, Rosemary E. Gale, Inés Gómez-Seguí, Giovani Marconi, Jean Norden, Jude Fitzgibbon, Mette K. Andersen, M Bornhäuser, Sarah E. Fordham, Amanda F. Gilkes, Heinz Sill, Eric A. Hungate, José Cervera, Friedrich Stölzel, Julia Gaal-Wesinger, Kim Piechocki, Wendy Stock, Theresa Hahn, Konstantin Strauch, David Allsup, Kenan Onel, Claire Elstob, Alyssa I. Clay-Gilmour, Nicola J. Sunter, Jelena D. Milosevic Feenstra, Meyling Cheok, Abrar Alharbi, Ann K. Daly, Sally Jeffries, Lisa Wagenführ, Olaf Heidenreich, Robert Kralovics, Alan K. Burnett, Giovanni Martinelli, Desiree Kunadt, Christian Gieger, Francesco Lo-Coco, Leo Ruhnke, Maria Teresa Voso, Junke Wang, Catherine Park, Nigel H. Russell, Chimène Moreilhon, Robert Kerrin Hills, Claude Preudhomme, Graham Jackson, Daniel Nowak, Maria Chiara Fontana, James M. Allan, Heidi Altmann, Richard S. Houlston, Anne S. Quante, Michelle M. Le Beau, Thahira Rahman, Christoph Röllig, Rebecca Darlay, Sophie Raynaud, Helen Marr, Csaba Bödör, Louise Palm, Thomas Cluzeau, Szilvia Krizsán, Heather J. Cordell, Mathew Collin, Torsten Haferlach, Lara E. Sucheston-Campbell, Wolf-Karsten Hofmann, Kimmo Porkka, Andras Masszi, Hervé Dombret, Miguel A. Sanz, Elisabeth Douglas, Tobias Menne, HUS Comprehensive Cancer Center, University Management, Helsinki University Hospital Area, Department of Oncology, and Hematologian yksikkö
- Subjects
Oncology ,General Physics and Astronomy ,Genome-wide association study ,Disease ,0302 clinical medicine ,AML ,HLA Antigens ,hemic and lymphatic diseases ,Histone methylation ,Cancer genomics ,RISK ,0303 health sciences ,Multidisciplinary ,Myeloid leukemia ,Middle Aged ,CLONAL EVOLUTION ,CANCER ,3. Good health ,HLA ,Leukemia, Myeloid, Acute ,030220 oncology & carcinogenesis ,GENE-MUTATIONS ,medicine.medical_specialty ,Genotype ,Science ,Locus (genetics) ,HIF-1-ALPHA ,Human leukocyte antigen ,Polymorphism, Single Nucleotide ,Article ,Acute myeloid leukaemia ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Aldehyde Reductase ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,030304 developmental biology ,Genetic association ,OLDER PATIENTS ,Whites ,business.industry ,HUMAN-LEUKOCYTE ANTIGEN ,Reproducibility of Results ,Cancer ,General Chemistry ,Settore MED/15 ,medicine.disease ,IMMUNE ESCAPE ,Risk factors ,Case-Control Studies ,3111 Biomedicine ,business ,Genome-Wide Association Study - Abstract
Acute myeloid leukemia (AML) is a hematological malignancy with an undefined heritable risk. Here we perform a meta-analysis of three genome-wide association studies, with replication in a fourth study, incorporating a total of 4018 AML cases and 10488 controls. We identify a genome-wide significant risk locus for AML at 11q13.2 (rs4930561; P = 2.15 × 10−8; KMT5B). We also identify a genome-wide significant risk locus for the cytogenetically normal AML sub-group (N = 1287) at 6p21.32 (rs3916765; P = 1.51 × 10−10; HLA). Our results inform on AML etiology and identify putative functional genes operating in histone methylation (KMT5B) and immune function (HLA)., Genome wide association studies in cancer are used to understand the heritable genetic contribution to disease risk. Here, the authors perform a genome wide association study in European patients with acute myeloid leukemia and identify loci associated with risk of developing the disease.
- Published
- 2021
16. Genome-wide association study identifies susceptibility loci for acute myeloid leukemia
- Author
-
Michelle M. Le Beau, Thahira Rahman, Yaobo Xu, Wendy Stock, Andrew D. Skol, Abrar Alharbi, David Allsup, Claire Elstob, Lara E. Sucheston-Campbell, Lisa Wagenführ, Olaf Heidenreich, Claude Preudhomme, Tobias Menne, Szilvia Krizsán, Rebecca Darlay, Jelena D. Milosevic Feenstra, David C. Linch, Sophie Raynaud, Helen Marr, Christian Gieger, Francesco Lo-Coco, David Grimwade, Maria Teresa Voso, Junke Wang, Christoph Röllig, Clare Lendrem, Wolf-Karsten Hofmann, Mathew Collin, Manja Meggendorfer, Friedrich Stölzel, Wei-Yu Lin, Ann K. Daly, Theresa Hahn, Torsten Haferlach, Sally Jeffries, Julia Gaal-Wesinger, Konstantin Strauch, Giovani Marconi, Amanda F. Gilkes, Chimène Moreilhon, Giovanni Martinelli, Anne M. Dickinson, Robert Kerrin Hills, Alan K. Burnett, Mette K. Andersen, Leo Ruhnke, Kimmo Porkka, Catherine Park, Desiree Kunadt, Nigel H. Russell, M Bornhäuser, Alyssa I. Clay-Gilmour, Hervé Dombret, Sarah E. Fordham, Eric A. Hungate, Miguel A. Sanz, Inés Gómez-Seguí, Csaba Bödör, Jean Norden, Elisabeth Douglas, Rosemary E. Gale, Heinz Sill, Kim Piechocki, Richard A. Larson, Robert Kralovics, Meyling Cheok, Heidi Altmann, Richard S. Houlston, Andras Masszi, Anne S. Quante, Louise Palm, Thomas Cluzeau, Heather J. Cordell, Nicola J. Sunter, Graham Jackson, Daniel Nowak, Maria Chiara Fontana, James M. Allan, José Cervera, Kenan Onel, Gail Jones, Adam Ivey, and Jude Fitzgibbon
- Subjects
0303 health sciences ,Myeloid leukemia ,Locus (genetics) ,Genome-wide association study ,Human leukocyte antigen ,Biology ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Histone methylation ,Cancer research ,Etiology ,030304 developmental biology ,Genetic association - Abstract
Acute myeloid leukemia (AML) is a hematological malignancy with an undefined heritable risk. Here we performed a meta-analysis of three genome-wide association studies, with replication in a fourth study, incorporating a total of 4018 AML cases and 10488 controls. We identified a genome-wide significant risk locus for AML at 11q13.2 (rs4930561; P = 2.15 × 10-8; KMT5B). We also identified a genome-wide significant risk locus for the cytogenetically normal AML sub-group (N=1287) at 6p21.32 (rs3916765; P = 1.51 × 10-10; HLA). Our results inform on AML etiology by identifying putative functional genes operating in histone methylation (KMT5B) and immune function (HLA).
- Published
- 2021
- Full Text
- View/download PDF
17. Integration of genomics and transcriptomics predicts diabetic retinopathy susceptibility genes
- Author
-
Segun Jung, Barbara E. Stranger, Maria Sverdlov, Dingcai Cao, Ionut Bebu, Amy Y. Lin, Ana Marija Sokovic, Michael A. Grassi, Dcct, Sarah Fazal, Andrew D Skol, Siquan Chen, Poulami P Borkar, Olukayode A. Sosina, and Anand Swaroop
- Subjects
Male ,0301 basic medicine ,folliculin ,Disease ,Bioinformatics ,Transcriptome ,0302 clinical medicine ,Lymphocytes ,Biology (General) ,Cell Line, Transformed ,General Neuroscience ,General Medicine ,Diabetic retinopathy ,diabetic retinopathy ,030220 oncology & carcinogenesis ,Medicine ,Female ,Research Article ,Human ,Retinopathy ,Adult ,QH301-705.5 ,Science ,Quantitative Trait Loci ,Blood sugar ,eQTL ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Young Adult ,03 medical and health sciences ,LCLs ,Proto-Oncogene Proteins ,Diabetes mellitus ,medicine ,Humans ,Genetic Predisposition to Disease ,Folliculin ,Gene ,General Immunology and Microbiology ,business.industry ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Genetics and Genomics ,Mendelian Randomization Analysis ,medicine.disease ,Diabetes Mellitus, Type 1 ,Glucose ,030104 developmental biology ,Case-Control Studies ,gene expression ,mendelian randomization ,business ,Genome-Wide Association Study - Abstract
We determined differential gene expression in response to high glucose in lymphoblastoid cell lines derived from matched individuals with type 1 diabetes with and without retinopathy. Those genes exhibiting the largest difference in glucose response were assessed for association with diabetic retinopathy in a genome-wide association study meta-analysis. Expression quantitative trait loci (eQTLs) of the glucose response genes were tested for association with diabetic retinopathy. We detected an enrichment of the eQTLs from the glucose response genes among small association p-values and identified folliculin (FLCN) as a susceptibility gene for diabetic retinopathy. Expression of FLCN in response to glucose was greater in individuals with diabetic retinopathy. Independent cohorts of individuals with diabetes revealed an association of FLCN eQTLs with diabetic retinopathy. Mendelian randomization confirmed a direct positive effect of increased FLCN expression on retinopathy. Integrating genetic association with gene expression implicated FLCN as a disease gene for diabetic retinopathy., eLife digest One of the side effects of diabetes is loss of vision from diabetic retinopathy, which is caused by injury to the light sensing tissue in the eye, the retina. Almost all individuals with diabetes develop diabetic retinopathy to some extent, and it is the leading cause of irreversible vision loss in working-age adults in the United States. How long a person has been living with diabetes, the extent of increased blood sugars and genetics all contribute to the risk and severity of diabetic retinopathy. Unfortunately, virtually no genes associated with diabetic retinopathy have yet been identified. When a gene is activated, it produces messenger molecules known as mRNA that are used by cells as instructions to produce proteins. The analysis of mRNA molecules, as well as genes themselves, can reveal the role of certain genes in disease. The studies of all genes and their associated mRNAs are respectively called genomics and transcriptomics. Genomics reveals what genes are present, while transcriptomics shows how active genes are in different cells. Skol et al. developed methods to study genomics and transcriptomics together to help discover genes that cause diabetic retinopathy. Genes involved in how cells respond to high blood sugar were first identified using cells grown in the lab. By comparing the activity of these genes in people with and without retinopathy the study identified genes associated with an increased risk of retinopathy in diabetes. In people with retinopathy, the activity of the folliculin gene (FLCN) increased more in response to high blood sugar. This was further verified with independent groups of people and using computer models to estimate the effect of different versions of the folliculin gene. The methods used here could be applied to understand complex genetics in other diseases. The results provide new understanding of the effects of diabetes. They may also help in the development of new treatments for diabetic retinopathy, which are likely to improve on the current approach of using laser surgery or injections into the eye.
- Published
- 2020
- Full Text
- View/download PDF
18. Author response: Integration of genomics and transcriptomics predicts diabetic retinopathy susceptibility genes
- Author
-
Maria Sverdlov, Ionut Bebu, Michael A. Grassi, Poulami P Borkar, Olukayode A. Sosina, Anand Swaroop, Ana Marija Sokovic, Barbara E. Stranger, Segun Jung, Siquan Chen, Andrew D Skol, Amy Y. Lin, Dingcai Cao, Sarah Fazal, and Dcct
- Subjects
Transcriptome ,medicine ,Genomics ,Susceptibility gene ,Computational biology ,Diabetic retinopathy ,Biology ,medicine.disease - Published
- 2020
- Full Text
- View/download PDF
19. The GTEx Consortium atlas of genetic regulatory effects across human tissues
- Author
-
Deborah C. Mash, Kevin S. Smith, Lappalainen T, Jeffrey A. Thomas, Rajinder Kaul, Paul Flicek, Maghboeba Mosavel, Yuxin Zou, Barbara E. Stranger, Brandon L. Pierce, Yanyu Liang, Andrew R. Hamel, Lihua Jiang, Marcus Hunter, Jimmie B. Vaught, Hae Kyung Im, John M. Rouhana, François Aguet, Ferran Reverter, Jason Bridge, Farzana Jasmine, Scott D. Jewell, William F. Leinweber, Gad Getz, Jonah Einson, Kevin Myer, SE Castel, Barbara E. Engelhardt, Stephen B. Montgomery, Brunilda Balliu, Gary Walters, Helen M. Moore, Daniel Nachun, Zerbino, Lori E. Brigham, Gao Wang, Farhad Hormozdiari, Pejman Mohammadi, Kasper D. Hansen, Nicole A. Teran, Fred A. Wright, Bryan Gillard, Sarah Kim-Hellmuth, CC Powell, Susan E. Koester, Wucher, Aaron Graubert, Duyen T. Nguyen, Shin Lin, Mike Moser, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Peter Hickey, Latarsha J. Carithers, Saboor Shad, Eric R. Gamazon, Jennifer A. Doherty, Stephen J. Trevanion, Kane Hadley, Kate R. Rosenbloom, Anita H. Undale, Robert E. Handsaker, Debra Bradbury, Shankara Anand, Meng Wang, David E. Tabor, Karna Robinson, S. Gabriel, Esti Yeger-Lotem, Kimberly Ramsey, Mary Barcus, Daniel G. MacArthur, Yuan He, Nancy Roche, Alvaro N. Barbeira, Ayellet V. Segrè, Dan Sheppard, Souvik Das, AR Little, Nathan S. Abell, Xiaoquan Wen, Elise D. Flynn, Nicole M. Ferraro, Hua Tang, Jared L. Nedzel, Jessica Wheeler, Abhi Rao, Meier, Thomas Juettemann, Sandra Linder, Bruce A. Roe, Daniel J. Cotter, David A. Davis, Christopher Johns, Lin Chen, Seva Kashin, Muhammad G. Kibriya, Ana Viñuela, Ellen Todres, Ashis Saha, Matthew Stephens, Chiara Sabatti, Manolis Kellis, Laura A. Siminoff, Phillip Branton, Xiao Li, Michael Snyder, Kathryn Demanelis, Gen Li, Barbara A. Foster, Leslie H. Sobin, Simona Volpi, Magali Ruffier, Christopher D. Brown, Ping Guan, Benjamin J. Strober, Alexis Battle, Michael J. Gloudemans, Silva Kasela, Manuel Muñoz-Aguirre, Ellen Karasik, OM deGoede, Roderic Guigó, Michael Washington, Alisa McDonald, Andrew A. Brown, Meritxell Oliva, Kieron Taylor, Nancy J. Cox, Daniel C. Rohrer, Paul J. Hoffman, Gene Kopen, Qin Li, Andrew D Skol, Rodrigo Bonazzola, Tiffany Eulalio, Mark H. Johnson, Laure Fresard, Lindsay F. Rizzardi, Abhiram Rao, T Krubit, W. J. Kent, Alan Kwong, Anna M. Smith, Pedro G. Ferreira, HM Gardiner, Andrew P. Feinberg, Rick Hasz, Lei Hou, Marta Melé, Andrew B. Nobel, Katherine H. Huang, Laura Barker, Maximilian Haeussler, Kristin G. Ardlie, Concepcion R. Nierras, Christopher Lee, Joshua M. Akey, Eskin E, Jeffrey McLean, Donald F. Conrad, Jin Billy Li, YoSon Park, Serghei Mangul, Emmanouil T. Dermitzakis, Brian Jo, D Garrido-Martin, and Barcelona Supercomputing Center
- Subjects
Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,0303 health sciences ,Linkage disequilibrium ,Multidisciplinary ,Genotype-Tissue Expression (GTEx) ,Atlas (topology) ,Human tissues ,Cancer susceptibility ,Diseases ,Genome-wide association study ,RNA-Seq ,Computational biology ,Biology ,Expressió gènica ,Human genetics ,03 medical and health sciences ,Tissues ,0302 clinical medicine ,Allelic heterogeneity ,Gene expression ,Genètica ,030217 neurology & neurosurgery ,030304 developmental biology ,Teixits (Histologia) - Abstract
The Genotype-Tissue Expression (GTEx) project dissects how genetic variation affects gene expression and splicing. Some human genetic variants affect the amount of RNA produced and the splicing of gene transcripts, crucial steps in development and maintaining a healthy individual. However, some of these changes only occur in a small number of tissues within the body. The Genotype-Tissue Expression (GTEx) project has been expanded over time, and, looking at the final data in version 8, Aguet et al. present a deep characterization of genetic associations and gene expression and splicing in 838 individuals over 49 tissues (see the Perspective by Wilson). This large study was able to characterize the details underlying many aspects of gene expression and provides a resource with which to better understand the fundamental molecular mechanisms of how genetic variants affect gene regulation and complex traits in humans. Science, this issue p. 1318; see also p. 1298 The Genotype-Tissue Expression (GTEx) project was established to characterize genetic effects on the transcriptome across human tissues and to link these regulatory mechanisms to trait and disease associations. Here, we present analyses of the version 8 data, examining 15,201 RNA-sequencing samples from 49 tissues of 838 postmortem donors. We comprehensively characterize genetic associations for gene expression and splicing in cis and trans, showing that regulatory associations are found for almost all genes, and describe the underlying molecular mechanisms and their contribution to allelic heterogeneity and pleiotropy of complex traits. Leveraging the large diversity of tissues, we provide insights into the tissue specificity of genetic effects and show that cell type composition is a key factor in understanding gene regulatory mechanisms in human tissues. We thank the donors and their families for their generous gifts of organ donation for transplantation and tissue donations for the GTEx research project; the Genomics Platform at the Broad Institute for data generation; J. Struewing for support and leadership of the GTEx project; M. Khan and C. Stolte for the illustrations in Fig. 1; and R. Do, D. Jordan, and M. Verbanck for providing GWAS pleiotropy scores. Funding: This work was supported by the Common Fund of the Office of the Director, U.S. National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, NIA, NIAID, and NINDS through NIH contracts HHSN261200800001E (Leidos Prime contract with NCI: A.M.S., D.E.T., N.V.R., J.A.M., L.S., M.E.B., L.Q., T.K., D.B., K.R., and A.U.), 10XS170 (NDRI: W.F.L., J.A.T., G.K., A.M., S.S., R.H., G.Wa., M.J., M.Wa., L.E.B., C.J., J.W., B.R., M.Hu., K.M., L.A.S., H.M.G., M.Mo., and L.K.B.), 10XS171 (Roswell Park Cancer Institute: B.A.F., M.T.M., E.K., B.M.G., K.D.R., and J.B.), 10X172 (Science Care Inc.), 12ST1039 (IDOX), 10ST1035 (Van Andel Institute: S.D.J., D.C.R., and D.R.V.), HHSN268201000029C (Broad Institute: F.A., G.G., K.G.A., A.V.S., X.Li., E.T., S.G., A.G., S.A., K.H.H., D.T.N., K.H., S.R.M., and J.L.N.), 5U41HG009494 (F.A., G.G., and K.G.A.), and through NIH grants R01 DA006227-17 (University of Miami Brain Bank: D.C.M. and D.A.D.), Supplement to University of Miami grant DA006227 (D.C.M. and D.A.D.), R01 MH090941 (University of Geneva), R01 MH090951 and R01 MH090937 (University of Chicago), R01 MH090936 (University of North Carolina–Chapel Hill), R01MH101814 (M.M.-A., V.W., S.B.M., R.G., E.T.D., D.G.-M., and A.V.), U01HG007593 (S.B.M.), R01MH101822 (C.D.B.), U01HG007598 (M.O. and B.E.S.), U01MH104393 (A.P.F.), extension H002371 to 5U41HG002371 (W.J.K.), as well as other funding sources: R01MH106842 (T.L., P.M., E.F., and P.J.H.), R01HL142028 (T.L., Si.Ka., and P.J.H.), R01GM122924 (T.L. and S.E.C.), R01MH107666 (H.K.I.), P30DK020595 (H.K.I.), UM1HG008901 (T.L.), R01GM124486 (T.L.), R01HG010067 (Y.Pa.), R01HG002585 (G.Wa. and M.St.), Gordon and Betty Moore Foundation GBMF 4559 (G.Wa. and M.St.), 1K99HG009916-01 (S.E.C.), R01HG006855 (Se.Ka. and R.E.H.), BIO2015-70777-P, Ministerio de Economia y Competitividad and FEDER funds (M.M.-A., V.W., R.G., and D.G.-M.), la Caixa Foundation ID 100010434 under agreement LCF/BQ/SO15/52260001 (D.G.-M.), NIH CTSA grant UL1TR002550-01 (P.M.), Marie-Skłodowska Curie fellowship H2020 Grant 706636 (S.K.-H.), R35HG010718 (E.R.G.), FPU15/03635, Ministerio de Educación, Cultura y Deporte (M.M.-A.),R01MH109905, 1R01HG010480 (A.Ba.), Searle Scholar Program (A.Ba.), R01HG008150 (S.B.M.), 5T32HG000044-22, NHGRI Institutional Training Grant in Genome Science (N.R.G.), EU IMI program (UE7-DIRECT-115317-1) (E.T.D. and A.V.), FNS funded project RNA1 (31003A_149984) (E.T.D. and A.V.), DK110919 (F.H.), F32HG009987 (F.H.), Massachusetts Lions Eye Research Fund Grant (A.R.H.), Wellcome grant WT108749/Z/15/Z (P.F.), and European Molecular Biology Laboratory (P.F. and D.Z.). Peer Reviewed "Article signat per 1 autors/es del BSC membres del THE GTEX CONSORTIUM: Marta Mele Messeguer"
- Published
- 2020
20. The impact of sex on gene expression across human tissues
- Author
-
Tuuli Lappalainen, Andrew D. Skol, Valentin Wucher, Ekaterina A. Khramtsova, Diego Garrido-Martín, Stephane E. Castel, Stephen B. Montgomery, Emmanouil T. Dermitzakis, Anthony Payne, Barbara E. Stranger, José Manuel Soria, Ferran Reverter, Hae Kyung Im, Daniel J. Cotter, Rodrigo Bonazzola, Brunilda Balliu, Brandon L. Pierce, Eric R. Gamazon, Alexis Battle, Pejman Mohammadi, Andrew R Hamel, Roderic Guigó, Ariel D. H. Gewirtz, Manuel Muñoz-Aguirre, Kristin G. Ardlie, Patrick Evans, Alvaro N. Barbeira, Yuxin Zou, Andrew A. Brown, Silva Kasela, Matthew Stephens, Barbara E. Engelhardt, Sarah Kim-Hellmuth, Angel Martinez-Perez, Ana Viñuela, Meritxell Oliva, Eleazar Eskin, Ayellet V. Segrè, Princy Parsana, and François Aguet
- Subjects
Male ,Single sex ,Quantitative Trait Loci ,Gene Expression ,Genome-wide association study ,Quantitative trait locus ,Biology ,Genoma humà ,Article ,Epigenesis, Genetic ,Transcriptome ,Cellular origin ,Sex Factors ,Gene expression ,Genetic variation ,Humans ,Disease ,Precision Medicine ,Promoter Regions, Genetic ,Gene ,Genetics ,Regulation of gene expression ,Chromosomes, Human, X ,Sex Characteristics ,Multidisciplinary ,Human genome ,Cromosomes sexuals ,Genetic Variation ,Sex chromosomes ,Phenotype ,Expressió gènica ,Gene Expression Regulation ,Organ Specificity ,Expression quantitative trait loci ,Female ,Genome-Wide Association Study - Abstract
Many complex human phenotypes exhibit sex-differentiated characteristics. However, the molecular mechanisms underlying these differences remain largely unknown. We generated a catalog of sex differences in gene expression and in the genetic regulation of gene expression across 44 human tissue sources surveyed by the Genotype-Tissue Expression project (GTEx, v8 release). We demonstrate that sex influences gene expression levels and cellular composition of tissue samples across the human body. A total of 37% of all genes exhibit sex-biased expression in at least one tissue. We identify cis expression quantitative trait loci (eQTLs) with sex-differentiated effects and characterize their cellular origin. By integrating sex-biased eQTLs with genome-wide association study data, we identify 58 gene-trait associations that are driven by genetic regulation of gene expression in a single sex. These findings provide an extensive characterization of sex differences in the human transcriptome and its genetic regulation. INTRODUCTION: Many complex human pheno-types, including diseases, exhibit sex-differentiated characteristics. These sex differences have been variously attributed to hormones, sex chromosomes, genotype × sex effects, differences in behavior, and differences in environmental exposures; however, their mechanisms and underlying biology remain largely unknown. The Genotype-Tissue Expression (GTEx) project provides an opportunity to investigate the prevalence and genetic mechanisms of sex differences in the human transcriptome by surveying many tissues that have not previously been characterized in this manner. RATIONALE: To characterize sex differences in the human transcriptome and its regulation, and to discover how sex and genetics interact to influence complex traits and disease, we generated a catalog of sex differences in gene expression and its genetic regulation across 44 human tissue sources surveyed by the GTEx project (v8 data release), analyzing 16,245 RNA-sequencing samples and genotypes of 838 adult individuals. We report sex differences in gene expression levels, tissue cell type composition, and cis expression quantitative trait loci (cis-eQTLs). To assess their impact, we integrated these results with gene function, transcription factor binding annotation, and genome-wide association study (GWAS) summary statistics of 87 GWASs. RESULTS: Sex effects on gene expression are ubiquitous (13,294 sex-biased genes across all tissues). However, these effects are small and largely tissue-specific. Genes with sex-differentiated expression are not primarily driven by tissue-specific gene expression and are involved in a diverse set of biological functions, such as drug and hormone response, embryonic development and tissue morphogenesis, fertilization, sexual reproduction and spermatogenesis, fat metabolism, cancer, and immune response. Whereas X-linked genes with higher expression in females suggest candidates for escape from X-chromosome inactivation, sex-biased expression of autosomal genes suggests hormone-related transcription factor regulation and a role for additional transcription factors, as well as sex-differentiated distribution of epigenetic marks, particularly histone H3 Lys(27) trimethylation (H3K27me3). Sex differences in the genetic regulation of gene expression are much less common (369 sex-biased eQTLs across all tissues) and are highly tissue-specific. We identified 58 gene-trait associations driven by genetic regulation of gene expression in a single sex. These include loci where sex-differentiated cell type abundances mediate genotype-phenotype associations, as well as loci where sex may play a more direct role in the underlying molecular mechanism of the association. For example, we identified a female-specific eQTL in liver for the hexokinase HKDC1 that influences glucose metabolism in pregnant females, which is subsequently reflected in the birth weight of the offspring. CONCLUSION: By integrating sex-aware analyses of GTEx data with gene function and transcription factor binding annotations, we describe tissue-specific and tissue-shared drivers and mechanisms contributing to sex differences in the human transcriptome and eQTLs. We discovered multiple sex-differentiated genetic effects on gene expression that colocalize with complex trait genetic associations, thereby facilitating the mechanistic interpretation of GWAS signals. Because the causative tissue is unknown for many phenotypes, analysis of the diverse GTEx tissue collection can serve as a powerful resource for investigations into the basis of sex-biased traits. This work provides an extensive characterization of sex differences in the human transcriptome and its genetic regulation. ■
- Published
- 2020
21. Mendelian randomization identifies folliculin expression as a mediator of diabetic retinopathy
- Author
-
Barbara E. Stranger, Maria Sverdlov, Ionut Bebu, Andrew D Skol, Amy Y. Lin, Michael A. Grassi, Segun Jung, Sarah Fazal, Siquan Chen, Poulami P Borkar, Dcct, Olukayode A. Sosina, Anand Swaroop, Dingcai Cao, and Ana Marija Sokovic
- Subjects
Type 1 diabetes ,business.industry ,Diabetes mellitus ,Mendelian randomization ,Expression quantitative trait loci ,Medicine ,Diabetic retinopathy ,Folliculin ,business ,medicine.disease ,Bioinformatics ,Genetic association ,Retinopathy - Abstract
The goal of the study was to identify genes whose aberrant expression can contribute to diabetic retinopathy. We determined differential gene expression in response to high glucose in lymphoblastoid cell lines derived from matched individuals with type 1 diabetes (T1D) with and without retinopathy. Those genes exhibiting the largest difference in glucose response between individuals with diabetes with and without retinopathy were assessed for association to diabetic retinopathy utilizing genotype data from a genome-wide association study meta-analysis. All genetic variants associated with gene expression (expression Quantitative Trait Loci, eQTLs) of the glucose response genes were tested for association with diabetic retinopathy. We detected an enrichment of the eQTLs from the glucose response genes among small association p-values and identified folliculin (FLCN) as a susceptibility gene for diabetic retinopathy. We show that expression of FLCN in response to glucose was greater in individuals with diabetic retinopathy compared to individuals with diabetes without retinopathy. Three large, independent cohorts of individuals with diabetes revealed an association of FLCN eQTLs to diabetic retinopathy. Mendelian randomization further confirmed a direct positive effect of increased FLCN expression on retinopathy in individuals with diabetes. Together, our studies integrating genetic association and gene expression implicate FLCN as a disease gene for diabetic retinopathy.
- Published
- 2020
- Full Text
- View/download PDF
22. Functional Common and Rare
- Author
-
Riyue, Bao, Anita, Ng, Mark, Sasaki, Myvizhi, Esai Selvan, Alyna, Katti, Hyesan, Lee, Lei, Huang, Andrew D, Skol, Cinzia, Lavarino, Hector, Salvador, Robert J, Klein, Zeynep H, Gümüş, Jaume, Mora, and Kenan, Onel
- Subjects
Adult ,Male ,Adolescent ,Receptor, ErbB-2 ,DNA Mutational Analysis ,Article ,Pedigree ,Gene Expression Regulation, Neoplastic ,Young Adult ,Neoplastic Syndromes, Hereditary ,Exome Sequencing ,Biomarkers, Tumor ,Humans ,Exome ,Female ,Genetic Predisposition to Disease ,Genetic Testing ,Child ,Germ-Line Mutation ,Aged - Abstract
We investigated a Spanish and Catalan family in which multiple cancer types tracked across three generations, but for which no genetic etiology had been identified. Whole exome sequencing of germline DNA from multiple affected family members was performed to identify candidate variants to explain this occurrence of familial cancer. We discovered in all cancer-affected family members a single rare heterozygous germline variant (I654V, rs1801201) in ERBB2/HER2 that is located in a transmembrane glycine zipper motif critical for ERBB2-mediated signaling and in complete linkage disequilibrium (D’=1) with a common polymorphism (I655V, rs1136201) previously reported in some populations as associated with cancer risk. Because multiple cancer types occurred in this family, we tested both the I654V and the I655V variants for association with cancer across multiple tumor types in 6,371 cases of Northern European ancestry drawn from The Cancer Genome Atlas and 6,647 controls, and found that the rare variant (I654V) was significantly associated with an increased risk for cancer (OR=1.40, p=0.021, 95% CI=1.05–1.89). Functional assays performed in HEK 293T cells revealed that both the I655V single mutant (SM) and the I654V;I655V double mutant (DM) stabilized ERBB2 protein and activated ERBB2 signaling, with the DM activating ERBB2 significantly more than the SM alone. Thus, our results suggest a model whereby heritable genetic variation in the transmembrane domain activating ERBB2 signaling is associated with both sporadic and familial cancer risk, with increased ERBB2 stabilization and activation associated with increased cancer risk.
- Published
- 2020
23. A vast resource of allelic expression data spanning human tissues
- Author
-
Nancy J. Cox, Sayantan Das, Abhi Rao, Pejman Mohammadi, Alan Kwong, Brandon L. Pierce, Yanyu Liang, Yuxin Zou, Anna M. Smith, Matthew Stephens, Chiara Sabatti, Yuan He, Kasper D. Hansen, Lei Hou, Meritxell Oliva, W. James Kent, Stacey Gabriel, Andrew R. Hame, Tanya Krubit, Gary Walters, Lori E. Brigham, Gao Wang, Kevin S. Smith, Michael J. Gloudemans, Barbara E. Engelhardt, Yongjin Park, Nicole A. Teran, David A. Davis, Thomas Juettemann, Kimberley Ramsey, Fred A. Wright, Lin Chen, Valentin Wucher, Benjamin J. Strober, Duyen T. Nguyen, Eleazar Eskin, Kane Hadley, Deborah C. Mash, Michael Snyder, Sarah Kim-Hellmuth, Laura A. Siminoff, Maghboeba Mosavel, Shin Lin, Richard Hasz, Daniel C. Rohrer, Latarsha J. Carithers, Kevin Myer, Rajinder Kaul, Andrew D. Skol, Bryan Gillard, Dana R. Valley, Philip A. Branton, Stephane E. Castel, Robert E. Handsaker, Debra Bradbury, Meng Wang, Mary Barcus, Xiaoquan Wen, Hua Tang, Daniel J. Cotter, Lihua Jiang, Jason Bridge, Ashis Saha, Gen Li, Susan E. Koester, Qin Li, Mark H. Johnson, Barbara E. Stranger, Jimmie B. Vaught, Hae Kyung Im, Paul Flicek, Marcus Hunter, François Aguet, Elise D. Flynn, Sandra Linder, Nancy Roche, Daniel R. Zerbino, Xiao Li, Barbara A. Foster, Stephen B. Montgomery, Daniel Nachun, Serghei Mangul, Emmanouil T. Dermitzakis, Brian Jo, Simona Volpi, Farzana Jasmine, Scott D. Jewell, Jonah Einson, Tuuli Lappalainen, Farhad Hormozdiari, John M. Rouhana, Ana Viñuela, Daniel G. MacArthur, William F. Leinweber, Gad Getz, Peter Hickey, Eric R. Gamazon, Brunilda Balliu, Jennifer A. Doherty, Christopher D. Brown, Roderic Guigó, Gene Kopen, Rodrigo Bonazzola, Pedro G. Ferreira, Andrew P. Feinberg, Shankara Anand, Helen M. Moore, Paul J. Hoffman, Heather M. Gardiner, Ping Guan, Ferran Reverter, Jin Billy Li, Tiffany Eulalio, Joseph Wheeler, Alvaro N. Barbeira, Jared L. Nedzel, Seva Kashin, Laure Fresard, Lindsay F. Rizzardi, Abhiram Rao, Muhammad G. Kibriya, David Tabor, Leslie H. Sobin, A. Roger Little, Stephen J. Trevanion, Nicole M. Ferraro, Kate R. Rosenbloom, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Ayellet V. Segrè, Dan Sheppard, Nathan S. Abell, Kathryn Demanelis, Manolis Kellis, Silva Kasela, Xin Li, Conner C. Powell, YoSon Park, Michael Washington, Magali Ruffier, Saboor Shad, Christopher Johns, Jeffrey A. Thomas, Andrew Brown, Alisa McDonald, Karna Robinson, Esti Yeger-Lotem, Manuel Muñoz-Aguirre, Kieron Taylor, Marta Melé, Diego Garrido-Martín, Brian Roe, Michael T. Moser, Andrew B. Nobel, Alexis Battle, Maximilian Haeussler, Concepcion R. Nierras, Ellen Karasik, Sam Meier, Anita H. Undale, Ellen Todres, Aaron Graubert, Joshua M. Akey, Jeffrey McLean, Donald F. Conrad, Olivia M. De Goede, Katherine H. Huang, Laura Barker, Kristin G. Ardlie, and Christopher Lee
- Subjects
animal structures ,Future studies ,lcsh:QH426-470 ,Short Report ,Gene Expression ,Genomics ,Computational biology ,Biology ,eQTL ,Polymorphism, Single Nucleotide ,ASE ,Regulatory variation ,03 medical and health sciences ,0302 clinical medicine ,Resource (project management) ,Humans ,SNP ,Allele ,lcsh:QH301-705.5 ,Alleles ,Allele specific ,030304 developmental biology ,0303 health sciences ,Allelic expression ,Genome, Human ,Sequence Analysis, RNA ,Haplotype ,Functional genomics ,Human genetics ,lcsh:Genetics ,Haplotypes ,lcsh:Biology (General) ,Expression data ,Expression quantitative trait loci ,GTEx ,030217 neurology & neurosurgery - Abstract
Allele expression (AE) analysis robustly measures cis-regulatory effects. Here, we present and demonstrate the utility of a vast AE resource generated from the GTEx v8 release, containing 15,253 samples spanning 54 human tissues for a total of 431 million measurements of AE at the SNP level and 153 million measurements at the haplotype level. In addition, we develop an extension of our tool phASER that allows effect sizes of cis-regulatory variants to be estimated using haplotype-level AE data. This AE resource is the largest to date, and we are able to make haplotype-level data publicly available. We anticipate that the availability of this resource will enable future studies of regulatory variation across human tissues.
- Published
- 2020
24. Correction to: PIK3R5 genetic predictors of hypertension induced by VEGF-pathway inhibitors
- Author
-
Julia C. F. Quintanilha, Alessandro Racioppi, Jin Wang, Amy S. Etheridge, Stefanie Denning, Carol E. Peña, Andrew D. Skol, Daniel J. Crona, Danyu Lin, and Federico Innocenti
- Subjects
Pharmacology ,Genetics ,Molecular Medicine - Published
- 2021
- Full Text
- View/download PDF
25. Does MAOA increase susceptibility to prenatal stress in young children?
- Author
-
Amalia E. Hatcher, Kimberly Andrews Espy, Daniel S. Pine, Edwin H. Cook, James L. Burns, Daniel K. Mroczek, Caron A. C. Clark, Lauren S. Wakschlag, Suena H. Massey, Andrew D. Skol, and David Goldman
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Physiology ,Toxicology ,Article ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Pregnancy ,Internal medicine ,Tobacco Smoking ,Genetic predisposition ,medicine ,Humans ,Genetic Predisposition to Disease ,Prospective Studies ,Young adult ,Child ,Prospective cohort study ,Monoamine Oxidase ,Problem Behavior ,Sex Characteristics ,biology ,medicine.disease ,030227 psychiatry ,Endocrinology ,Prenatal stress ,Child, Preschool ,Prenatal Exposure Delayed Effects ,Cohort ,biology.protein ,Female ,Gene-Environment Interaction ,Monoamine oxidase A ,Psychology ,Stress, Psychological ,030217 neurology & neurosurgery ,Sex characteristics - Abstract
Background We previously demonstrated a gene-by-prenatal-environment interaction whereby the monoamine oxidase A gene (MAOA) modified the impact of prenatal tobacco exposure (PTE) on adolescent disruptive behavior (DB), with the MAOA risk genotype varying by sex. We extend this work by examining whether this mechanism is evident with another common adversity, prenatal stress exposure (PSE), and whether sex differences are present earlier in development in closer proximity to exposure. Methods Participants were 281 mothers and their 285 children derived from a prenatal cohort with in-depth prospective measures of PSE and PTE. We assessed DB at age 5 via dimensional developmentally-sensitive measurement. Analyses were stratified by sex based on prior evidence for sex differences. Results Concurrent stress exposure predicted DB in children (β = 0.310, p = 0.001), while main effects of prenatal exposures were seen only in boys. We found a three-way interaction of MAOA × PSE × sex on DB (β = 0.813, p = 0.022). Boys with MAOA-H had more DB as a function of PSE, controlling for PTE (β = 0.774, p = 0.015), and as a function of PTE, controlling for PSE (β = 0.362, p = 0.037). Boys with MAOA-L did not show this susceptibility. MAOA did not interact with PSE (β = − 0.133, p = 0.561) nor PTE (β = − 0.144; p = 0.505) in predicting DB in girls. Examination of gene-environment correlation (rGE) showed a correlation between paternal MAOA-L and daughters' concurrent stress exposure (r = − 0.240, p = 0.013). Discussion Findings underscore complex mechanisms linking genetic susceptibility and early adverse exposures. Replication in larger cohorts followed from the pregnancy through adolescence is suggested to elucidate mechanisms that appear to have varying developmental expression.
- Published
- 2017
- Full Text
- View/download PDF
26. Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib
- Author
-
Andrew D. Skol, Nancy Klauber-DeMore, Dylan M. Glubb, Yuri K. Peterson, Kari Alitalo, Amy S. Etheridge, Daniel J. Crona, Veli-Matti Leppänen, Carol Peña, Eleanor Hilliard, Federico Innocenti, Kari Alitalo / Principal Investigator, University of Helsinki, HUSLAB, and CAN-PRO - Translational Cancer Medicine Program
- Subjects
0301 basic medicine ,Sorafenib ,Cancer Research ,endocrine system ,Angiogenesis ,3122 Cancers ,urologic and male genital diseases ,ANGIOGENESIS ,MECHANISMS ,ACTIVATION ,03 medical and health sciences ,0302 clinical medicine ,MULTIKINASE INHIBITOR ,Renal cell carcinoma ,Medicine ,Survival rate ,ITGAV ,Tube formation ,business.industry ,Cancer ,ASSOCIATION ,medicine.disease ,CANCER ,3. Good health ,Vascular endothelial growth factor A ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,LIGAND-BINDING ,Cancer research ,GROWTH ,3111 Biomedicine ,business ,FACTOR-TARGETED THERAPY ,medicine.drug - Abstract
Molecular markers of sorafenib efficacy in patients with metastatic renal cell carcinoma (mRCC) are not available. The purpose of this study was to discover genetic markers of survival in patients with mRCC treated with sorafenib. Germline variants from 56 genes were genotyped in 295 patients with mRCC. Variant-overall survival (OS) associations were tested in multivariate regression models. Mechanistic studies were conducted to validate clinical associations. VEGFA rs1885657, ITGAV rs3816375, and WWOX rs8047917 (sorafenib arm), and FLT4 rs307826 and VEGFA rs3024987 (sorafenib and placebo arms combined) were associated with shorter OS. FLT4 rs307826 increased VEGFR-3 phosphorylation, membrane trafficking, and receptor activation. VEGFA rs1885657 and rs58159269 increased transcriptional activity of the constructs containing these variants in endothelial and RCC cell lines, and VEGFA rs58159269 increased endothelial cell proliferation and tube formation. FLT4 rs307826 and VEGFA rs58159269 led to reduced sorafenib cytotoxicity. Genetic variation in VEGFA and FLT4 could affect survival in sorafenib-treated patients with mRCC. These markers should be examined in additional malignancies treated with sorafenib and in other angiogenesis inhibitors used in mRCC. Significance: Clinical and mechanistic data identify germline genetic variants in VEGFA and FLT4 as markers of survival in patients with metastatic renal cell carcinoma.
- Published
- 2019
27. Population-scale tissue transcriptomics maps long non-coding RNAs to complex disease
- Author
-
Olivia M. de Goede, Daniel C. Nachun, Nicole M. Ferraro, Michael J. Gloudemans, Abhiram S. Rao, Craig Smail, Tiffany Y. Eulalio, François Aguet, Bernard Ng, Jishu Xu, Alvaro N. Barbeira, Stephane E. Castel, Sarah Kim-Hellmuth, YoSon Park, Alexandra J. Scott, Benjamin J. Strober, Christopher D. Brown, Xiaoquan Wen, Ira M. Hall, Alexis Battle, Tuuli Lappalainen, Hae Kyung Im, Kristin G. Ardlie, Sara Mostafavi, Thomas Quertermous, Karla Kirkegaard, Stephen B. Montgomery, Shankara Anand, Stacey Gabriel, Gad A. Getz, Aaron Graubert, Kane Hadley, Robert E. Handsaker, Katherine H. Huang, Xiao Li, Daniel G. MacArthur, Samuel R. Meier, Jared L. Nedzel, Duyen T. Nguyen, Ayellet V. Segrè, Ellen Todres, Brunilda Balliu, Rodrigo Bonazzola, Andrew Brown, Donald F. Conrad, Daniel J. Cotter, Nancy Cox, Sayantan Das, Emmanouil T. Dermitzakis, Jonah Einson, Barbara E. Engelhardt, Eleazar Eskin, Elise D. Flynn, Laure Fresard, Eric R. Gamazon, Diego Garrido-Martín, Nicole R. Gay, Roderic Guigó, Andrew R. Hamel, Yuan He, Paul J. Hoffman, Farhad Hormozdiari, Lei Hou, Brian Jo, Silva Kasela, Seva Kashin, Manolis Kellis, Alan Kwong, Xin Li, Yanyu Liang, Serghei Mangul, Pejman Mohammadi, Manuel Muñoz-Aguirre, Andrew B. Nobel, Meritxell Oliva, Yongjin Park, Princy Parsana, Ferran Reverter, John M. Rouhana, Chiara Sabatti, Ashis Saha, Matthew Stephens, Barbara E. Stranger, Nicole A. Teran, Ana Viñuela, Gao Wang, Fred Wright, Valentin Wucher, Yuxin Zou, Pedro G. Ferreira, Gen Li, Marta Melé, Esti Yeger-Lotem, Debra Bradbury, Tanya Krubit, Jeffrey A. McLean, Liqun Qi, Karna Robinson, Nancy V. Roche, Anna M. Smith, David E. Tabor, Anita Undale, Jason Bridge, Lori E. Brigham, Barbara A. Foster, Bryan M. Gillard, Richard Hasz, Marcus Hunter, Christopher Johns, Mark Johnson, Ellen Karasik, Gene Kopen, William F. Leinweber, Alisa McDonald, Michael T. Moser, Kevin Myer, Kimberley D. Ramsey, Brian Roe, Saboor Shad, Jeffrey A. Thomas, Gary Walters, Michael Washington, Joseph Wheeler, Scott D. Jewell, Daniel C. Rohrer, Dana R. Valley, David A. Davis, Deborah C. Mash, Mary E. Barcus, Philip A. Branton, Leslie Sobin, Laura K. Barker, Heather M. Gardiner, Maghboeba Mosavel, Laura A. Siminoff, Paul Flicek, Maximilian Haeussler, Thomas Juettemann, W. James Kent, Christopher M. Lee, Conner C. Powell, Kate R. Rosenbloom, Magali Ruffier, Dan Sheppard, Kieron Taylor, Stephen J. Trevanion, Daniel R. Zerbino, Nathan S. Abell, Joshua Akey, Lin Chen, Kathryn Demanelis, Jennifer A. Doherty, Andrew P. Feinberg, Kasper D. Hansen, Peter F. Hickey, Farzana Jasmine, Lihua Jiang, Rajinder Kaul, Muhammad G. Kibriya, Jin Billy Li, Qin Li, Shin Lin, Sandra E. Linder, Brandon L. Pierce, Lindsay F. Rizzardi, Andrew D. Skol, Kevin S. Smith, Michael Snyder, John Stamatoyannopoulos, Hua Tang, Meng Wang, Latarsha J. Carithers, Ping Guan, Susan E. Koester, A. Roger Little, Helen M. Moore, Concepcion R. Nierras, Abhi K. Rao, Jimmie B. Vaught, and Simona Volpi
- Subjects
Multifactorial Inheritance ,Population ,Quantitative Trait Loci ,Coronary Artery Disease ,Disease ,Computational biology ,Quantitative trait locus ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Humans ,education ,Gene ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Gene Expression Profiling ,Genetic Variation ,Inflammatory Bowel Diseases ,Long non-coding RNA ,Diabetes Mellitus, Type 1 ,Diabetes Mellitus, Type 2 ,Organ Specificity ,Expression quantitative trait loci ,Trait ,RNA, Long Noncoding ,030217 neurology & neurosurgery - Abstract
Long non-coding RNA (lncRNA) genes have well-established and important impacts on molecular and cellular functions. However, among the thousands of lncRNA genes, it is still a major challenge to identify the subset with disease or trait relevance. To systematically characterize these lncRNA genes, we used Genotype Tissue Expression (GTEx) project v8 genetic and multi-tissue transcriptomic data to profile the expression, genetic regulation, cellular contexts, and trait associations of 14,100 lncRNA genes across 49 tissues for 101 distinct complex genetic traits. Using these approaches, we identified 1,432 lncRNA gene-trait associations, 800 of which were not explained by stronger effects of neighboring protein-coding genes. This included associations between lncRNA quantitative trait loci and inflammatory bowel disease, type 1 and type 2 diabetes, and coronary artery disease, as well as rare variant associations to body mass index.
- Published
- 2021
- Full Text
- View/download PDF
28. A Quantitative Proteome Map of the Human Body
- Author
-
Lihua Jiang, Meng Wang, Shin Lin, Ruiqi Jian, Xiao Li, Joanne Chan, Guanlan Dong, Huaying Fang, Aaron E. Robinson, Michael P. Snyder, François Aguet, Shankara Anand, Kristin G. Ardlie, Stacey Gabriel, Gad Getz, Aaron Graubert, Kane Hadley, Robert E. Handsaker, Katherine H. Huang, Seva Kashin, Daniel G. MacArthur, Samuel R. Meier, Jared L. Nedzel, Duyen Y. Nguyen, Ayellet V. Segrè, Ellen Todres, Brunilda Balliu, Alvaro N. Barbeira, Alexis Battle, Rodrigo Bonazzola, Andrew Brown, Christopher D. Brown, Stephane E. Castel, Don Conrad, Daniel J. Cotter, Nancy Cox, Sayantan Das, Olivia M. de Goede, Emmanouil T. Dermitzakis, Barbara E. Engelhardt, Eleazar Eskin, Tiffany Y. Eulalio, Nicole M. Ferraro, Elise Flynn, Laure Fresard, Eric R. Gamazon, Diego Garrido-Martín, Nicole R. Gay, Roderic Guigó, Andrew R. Hamel, Yuan He, Paul J. Hoffman, Farhad Hormozdiari, Lei Hou, Hae Kyung Im, Brian Jo, Silva Kasela, Manolis Kellis, Sarah Kim-Hellmuth, Alan Kwong, Tuuli Lappalainen, Xin Li, Yanyu Liang, Serghei Mangul, Pejman Mohammadi, Stephen B. Montgomery, Manuel Muñoz-Aguirre, Daniel C. Nachun, Andrew B. Nobel, Meritxell Oliva, YoSon Park, Yongjin Park, Princy Parsana, Ferran Reverter, John M. Rouhana, Chiara Sabatti, Ashis Saha, Andrew D. Skol, Matthew Stephens, Barbara E. Stranger, Benjamin J. Strober, Nicole A. Teran, Ana Viñuela, Gao Wang, Xiaoquan Wen, Fred Wright, Valentin Wucher, Yuxin Zou, Pedro G. Ferreira, Gen Li, Marta Melé, Esti Yeger-Lotem, Mary E. Barcus, Debra Bradbury, Tanya Krubit, Jeffrey A. McLean, Liqun Qi, Karna Robinson, Nancy V. Roche, Anna M. Smith, Leslie Sobin, David E. Tabor, Anita Undale, Jason Bridge, Lori E. Brigham, Barbara A. Foster, Bryan M. Gillard, Richard Hasz, Marcus Hunter, Christopher Johns, Mark Johnson, Ellen Karasik, Gene Kopen, William F. Leinweber, Alisa McDonald, Michael T. Moser, Kevin Myer, Kimberley D. Ramsey, Brian Roe, Saboor Shad, Jeffrey A. Thomas, Gary Walters, Michael Washington, Joseph Wheeler, Scott D. Jewell, Daniel C. Rohrer, Dana R. Valley, David A. Davis, Deborah C. Mash, Philip A. Branton, Laura K. Barker, Heather M. Gardiner, Maghboeba Mosavel, Laura A. Siminoff, Paul Flicek, Maximilian Haeussler, Thomas Juettemann, W. James Kent, Christopher M. Lee, Conner C. Powell, Kate R. Rosenbloom, Magali Ruffier, Dan Sheppard, Kieron Taylor, Stephen J. Trevanion, Daniel R. Zerbino, Nathan S. Abell, Joshua Akey, Lin Chen, Kathryn Demanelis, Jennifer A. Doherty, Andrew P. Feinberg, Kasper D. Hansen, Peter F. Hickey, Farzana Jasmine, Rajinder Kaul, Muhammad G. Kibriya, Jin Billy Li, Qin Li, Sandra E. Linder, Brandon L. Pierce, Lindsay F. Rizzardi, Kevin S. Smith, John Stamatoyannopoulos, Hua Tang, Latarsha J. Carithers, Ping Guan, Susan E. Koester, A. Roger Little, Helen M. Moore, Concepcion R. Nierras, Abhi K. Rao, Jimmie B. Vaught, and Simona Volpi
- Subjects
Proteomics ,Cell signaling ,Proteome ,Quantitative proteomics ,Gene Expression ,Disease ,Computational biology ,Biology ,ENCODE ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Secretion ,RNA, Messenger ,Gene ,030304 developmental biology ,0303 health sciences ,Gene Expression Profiling ,RNA ,Metabolism ,Phenotype ,Secretory protein ,030217 neurology & neurosurgery - Abstract
Determining protein levels in each tissue and how they compare with RNA levels is important for understanding human biology and disease as well as regulatory processes that control protein levels. We quantified the relative protein levels from 12,627 genes across 32 normal human tissue types prepared by the GTEx project. Known and new tissue specific or enriched proteins (5,499) were identified and compared to transcriptome data. Many ubiquitous transcripts are found to encode highly tissue specific proteins. Discordance in the sites of RNA expression and protein detection also revealed potential sites of synthesis and action of protein signaling molecules. Overall, these results provide an extraordinary resource, and demonstrate that understanding protein levels can provide insights into metabolism, regulation, secretome, and human diseases.SummaryQuantitative proteome study of 32 human tissues and integrated analysis with transcriptome data revealed that understanding protein levels could provide in-depth knowledge to post transcriptional or translational regulations, human metabolism, secretome, and diseases.
- Published
- 2020
- Full Text
- View/download PDF
29. 24. Clinical considerations for migration between genome assemblies: Lessons learned in moving to GRCh38
- Author
-
Sabah Kadri and Andrew D Skol
- Subjects
Cancer Research ,Genetics ,Computational biology ,Biology ,Molecular Biology ,Genome - Published
- 2020
- Full Text
- View/download PDF
30. Genetic Variants of
- Author
-
Daniel J, Crona, Andrew D, Skol, Veli-Matti, Leppänen, Dylan M, Glubb, Amy S, Etheridge, Eleanor, Hilliard, Carol E, Peña, Yuri K, Peterson, Nancy, Klauber-DeMore, Kari K, Alitalo, and Federico, Innocenti
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,endocrine system ,Antineoplastic Agents ,Apoptosis ,urologic and male genital diseases ,Article ,Young Adult ,Double-Blind Method ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,neoplasms ,Carcinoma, Renal Cell ,Aged ,Cell Proliferation ,Aged, 80 and over ,Middle Aged ,Sorafenib ,Prognosis ,Vascular Endothelial Growth Factor Receptor-3 ,digestive system diseases ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Mutation ,Female ,Follow-Up Studies - Abstract
Molecular markers of sorafenib efficacy in metastatic renal cell carcinoma (mRCC) patients are not available. The purpose of this study was to discover genetic markers of survival in mRCC patients treated with sorafenib. Germline variants from 56 genes were genotyped in 295 mRCC patients. Variant-overall survival (OS) associations were tested in multivariate regression models. Mechanistic studies were conducted to validate clinical associations. VEGFA rs1885657, ITGAV rs3816375, and WWOX rs8047917 (sorafenib arm), and FLT4 rs307826 and VEGFA rs3024987 (sorafenib and placebo arms combined) were associated with shorter OS. FLT4 rs307826 increased VEGFR-3 phosphorylation, membrane trafficking and receptor activation. VEGFA rs1885657 and rs58159269 increased transcriptional activity of the constructs containing these variants in endothelial and RCC cell lines, and VEGFA rs58159269 increased endothelial cell proliferation and tube formation. FLT4 rs307826 and VEGFA rs58159269 led to reduced sorafenib cytotoxicity. Genetic variation in VEGFA and FLT4 could affect survival in sorafenib-treated mRCC patients. These markers should be examined in additional malignancies treated with sorafenib, and in other angiogenesis inhibitors used in mRCC.
- Published
- 2018
31. Co-expression networks reveal the tissue-specific regulation of transcription and splicing
- Author
-
Morgan Diegel, Laure Fresard, Lindsay F. Rizzardi, Yuan He, Monkol Lek, Daniel C. Rohrer, Boxiang Liu, Maximilian Haeussler, Heather M. Traino, Concepcion R. Nierras, Joseph Wheeler, Serghei Mangul, Fan Wu, Hualin S. Xi, Andrew D. Skol, Steven Hunter, Yaping Liu, Casandra A. Trowbridge, Brandon L. Pierce, Daniel Bates, Peter Hickey, Susan E. Koester, Bryan Gillard, Eric R. Gamazon, Jennifer A. Doherty, Jared L. Nedzel, Eric Haugen, Lori E. Brigham, Gao Wang, Dana R. Valley, Zachary Zappala, Emmanouil T. Dermitzakis, Seva Kashin, Ira M. Hall, John Vivian, Philip A. Branton, Barbara E. Stranger, Magali Ruffier, Melina Claussnitzer, Nancy Roche, Michael Washington, Halit Ongen, Brian Jo, Rachna Kumar, Jean Monlong, Yi-Hui Zhou, Kristen Lee, Stephane E. Castel, Mark Miklos, Alisa McDonald, Diego Garrido-Martín, Jimmie B. Vaught, Hae Kyung Im, Leslie H. Sobin, John T. Lonsdale, Audra K. Johnson, Rui Zhang, Nancy J. Cox, Christopher D. Brown, Paul Flicek, Ferran Reverter, Roderic Guigó, Tuuli Lappalainen, Sarah E. Gould, Deborah C. Mash, Michael T. Moser, Andrew B. Nobel, Takunda Matose, Jingchun Zhu, Joe R. Davis, Andrey A. Shabalin, Jie Quan, Pedro G. Ferreira, Taru Tukiainen, Ellen Gelfand, Cédric Howald, Buhm Han, Emily K. Tsang, Andrew P. Feinberg, Caroline Linke, Kane Hadley, Richard Sandstrom, Mark D. Johnson, Joshua M. Akey, Ian C. McDowell, Daniel R. Zerbino, Alexis Battle, Brian Roe, Daniel G. MacArthur, Ellen Karasik, Marcus Hunter, Anjené M. Addington, Thomas Juettemann, Konrad J. Karczewski, Duyen T. Nguyen, Lei Hou, Stephen B. Montgomery, YoSon Park, Nicole C. Lockart, Lin Chen, Rajinder Kaul, Ruiqi Jian, Robert G. Montroy, Xiao Li, Michael Snyder, Beryl B. Cummings, Kimberly M. Valentino, Ariel D. H. Gewirtz, François Aguet, Jeffrey McLean, Gary Walters, Farhad Hormozdiari, William F. Leinweber, Gad Getz, Jeffery P. Struewing, Anne Ndungu, Dan L. Nicolae, Benoit Molinie, Lihua Jiang, Michael Sammeth, W. James Kent, John Palowitch, Brian Craft, Donald F. Conrad, Kathryn Demanelis, Jason Bridge, Jin Billy Li, A. Roger Little, Nicholas Van Wittenberghe, Stephen J. Trevanion, Pejman Mohammadi, Michael S. Noble, Kate R. Rosenbloom, Marian S. Fernando, Benjamin J. Strober, Ping Guan, Brunilda Balliu, Yungil Kim, Kevin Myer, Christine B. Peterson, Pushpa Hariharan, Jae Hoon Sul, Abhi Rao, Michael F. Salvatore, Qin Li, Eun Yong Kang, Matthew T. Maurano, Ayellet V. Segrè, Dan Sheppard, Fred A. Wright, Matthew Stephens, Kasper D. Hansen, Chiara Sabatti, Kevin S. Smith, Xin Li, Ruth Barshir, Muhammad G. Kibriya, Farhan N. Damani, Manolis Kellis, Olivier Delaneau, Shin Lin, Richard Hasz, Michael J. Gloudemans, Anita H. Undale, Mary Goldman, Fidencio J. Neri, Katherine H. Huang, David E. Tabor, Manuel Muñoz-Aguirre, Maghboeba Mosavel, Simona Volpi, Latarsha J. Carithers, Anna M. Smith, Genna Gliner, Eleazar Eskin, Nikolaos I Panousis, Benedict Paten, Andrew A. Brown, Jessica Lin, Kieron Taylor, Robert E. Handsaker, Laura Barker, Casey Martin, Meng Wang, Farzana Jasmine, Scott D. Jewell, Nathan S. Abell, Kristin G. Ardlie, Shilpi Singh, Mary Barcus, Anthony Payne, Christopher Lee, Xiaoquan Wen, Nicola J. Rinaldi, Hua Tang, Yongjin Park, Christopher Johns, Saboor Shad, Judith B. Zaugg, Reza Sodaei, Maria M. Tomaszewski, David A. Davis, Joanne Chan, Laura A. Siminoff, Mark I. McCarthy, Ki Sung Um, Karna Robinson, Esti Yeger-Lotem, Martijn van de Bunt, Meritxell Oliva, Jemma Nelson, Negin Vatanian, Colby Chiang, Jeffrey A. Thomas, Alexandra J. Scott, Omer Basha, Jessica Halow, Panagiotis Papasaikas, Barbara A. Foster, Barbara E. Engelhardt, Sarah Kim-Hellmuth, Li Wang, Gireesh K. Bogu, Sandra Linder, Sarah Urbut, Ashis Saha, Gen Li, Bernadette Mestichelli, Chuan Gao, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Helen M. Moore, Gene Kopen, and GTEx, Consortium
- Subjects
Gene isoform ,0301 basic medicine ,Genotyping Techniques ,Bioinformatics ,RNA Splicing ,1.1 Normal biological development and functioning ,Gene regulatory network ,Method ,Genomics ,Computational biology ,Biology ,Medical and Health Sciences ,GTEx Consortium ,Transcriptome ,03 medical and health sciences ,Databases ,0302 clinical medicine ,Genetic ,Transcription (biology) ,Underpinning research ,Genetic variation ,Gene expression ,Genetics ,Humans ,ddc:576.5 ,Gene Regulatory Networks ,Polymorphism ,Gene ,Genetics (clinical) ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Gene Expression Profiling ,Human Genome ,Bayes Theorem ,Single Nucleotide ,Biological Sciences ,Gene expression profiling ,030104 developmental biology ,Gene Expression Regulation ,Organ Specificity ,RNA splicing ,RNA ,Generic health relevance ,Sequence Analysis ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Gene co-expression networks capture biologically important patterns in gene expression data, enabling functional analyses of genes, discovery of biomarkers, and interpretation of regulatory genetic variants. Most network analyses to date have been limited to assessing correlation between total gene expression levels in a single or small sets of tissues. Here, we have reconstructed networks that capture a much more complete set of regulatory relationships, specifically including regulation of relative isoform abundance and splicing, and tissue-specific connections unique to each of a diverse set of tissues. Using the Genotype-Tissue Expression (GTEx) project v6 RNA-sequencing data across 44 tissues in 449 individuals, we evaluated shared and tissue-specific network relationships. First, we developed a framework called Transcriptome Wide Networks (TWNs) for combining total expression and relative isoform levels into a single sparse network, capturing the complex interplay between the regulation of splicing and transcription. We built TWNs for sixteen tissues, and found that hubs with isoform node neighbors in these networks were strongly enriched for splicing and RNA binding genes, demonstrating their utility in unraveling regulation of splicing in the human transcriptome, and providing a set of candidate shared and tissue-specific regulatory hub genes. Next, we used a Bayesian biclustering model that identifies network edges between genes with co-expression in a single tissue to reconstruct tissue-specific networks (TSNs) for 27 distinct GTEx tissues and for four subsets of related tissues. Using both TWNs and TSNs, we characterized gene co-expression patterns shared across tissues. Finally, we found genetic variants associated with multiple neighboring nodes in our networks, supporting the estimated network structures and identifying 33 genetic variants with distant regulatory impact on transcription and splicing. Our networks provide an improved understanding of the complex relationships between genes in the human transcriptome, including tissue-specificity of gene co-expression, regulation of splicing, and the coordinated impact of genetic variation on transcription.
- Published
- 2017
32. Genetic association with B-cell acute lymphoblastic leukemia in allogeneic transplant patients differs by age and sex
- Author
-
Stephen R. Spellman, Ezgi Karaesmen, David Ellinghaus, Lara E. Sucheston-Campbell, Marcelo C. Pasquini, Alyssa I. Clay-Gilmour, David Tritchler, Anne Marie W. Block, Eric A. Hungate, Sheila N.J. Sait, Christopher A. Haiman, Qian Liu, Eva Ellinghaus, Loreall Pooler, Sebastiano Battaglia, Jacqueline Clavel, Theresa Hahn, Laurent Orsi, Qiang Hu, Daniel J. Weisdorf, Philip L. McCarthy, Xiaochun Zhu, Andrew D. Skol, Martin Stanulla, Daniel O. Stram, Kenan Onel, Leah Preus, Abbas Rizvi, Qianqian Zhu, Song Liu, Xin Sheng, Li Yan, and Christine B. Ambrosone
- Subjects
0301 basic medicine ,Genetics ,medicine.medical_specialty ,Lymphoid Neoplasia ,business.industry ,Mortality rate ,Incidence (epidemiology) ,Genome-wide association study ,Hematology ,Odds ratio ,Confidence interval ,03 medical and health sciences ,030104 developmental biology ,Internal medicine ,medicine ,Genetic predisposition ,Age of onset ,business ,Genetic association - Abstract
The incidence and mortality rates of B-cell acute lymphoblastic leukemia (B-ALL) differ by age and sex. To determine if inherited genetic susceptibility contributes to these differences we performed 2 genome-wide association studies (GWAS) by age, sex, and subtype and subsequent meta-analyses. The GWAS included 446 B-ALL cases, and 3027 healthy unrelated blood and marrow transplant (BMT) donors as controls from the Determining the Influence of Susceptibility Conveying Variants Related to One-Year Mortality after BMT (DISCOVeRY-BMT) study. We identified 1 novel variant, rs189434316, significantly associated with odds of normal cytogenetic B-ALL (odds ratio from meta-analysis [ORmeta] = 3.7; 95% confidence interval [CI], 2.5, 6.2; P value from meta-analysis [Pmeta] = 6.0 × 10-9). The previously reported pediatric B-ALL GWAS variant, rs11980379 (IKZF1), replicated in B-ALL pediatric patients (ORmeta = 2.3; 95% CI, 1.5, 3.7; Pmeta = 1.0 × 10-9), with evidence of heterogeneity (P = .02) between males and females. Sex differences in single-nucleotide polymorphism effect were seen in those >15 years (OR = 1.7; 95% CI, 1.4, 2.2, PMales = 6.38 × 10-6/OR = 1.1; 95% CI, 0.8, 1.5; PFemales = .6) but not ≤15 years (OR = 2.3; 95% CI, 1.4, 3.8; PMales = .0007/OR = 1.9; 95% CI, 1.2, 3.2; PFemales = .007). The latter association replicated in independent pediatric B-ALL cohorts. A previously identified adolescent and young-adult onset ALL-associated variant in GATA3 is associated with B-ALL risk in those >40 years. Our findings provide more evidence of the influence of genetics on B-ALL age of onset and we have shown the first evidence that IKZF1 associations with B-ALL may be sex and age specific.
- Published
- 2017
33. Radiation-induced apoptosis varies among individuals and is modified by sex and age
- Author
-
Gareth L. Bond, Andrew D. Skol, Elisabeth E. Bond, Michael Overholtzer, Kenan Onel, and Mark A. Applebaum
- Subjects
Adult ,Male ,Aging ,Physiology ,Apoptosis ,Biology ,Radiation Dosage ,Models, Biological ,Peripheral blood mononuclear cell ,Article ,Young Adult ,Sex Factors ,Reference Values ,Sex factors ,Healthy volunteers ,Humans ,Computer Simulation ,Radiology, Nuclear Medicine and imaging ,Young adult ,Cells, Cultured ,Radiation induced apoptosis ,Radiological and Ultrasound Technology ,Middle Aged ,In vitro ,Immunology ,Leukocytes, Mononuclear ,Female ,Functional variation - Abstract
PURPOSE: Although there are considerable data on mechanisms of radiation-induced apoptosis in vitro and in animal models, little is known about functional variation in these pathways in humans. We sought to develop a tractable system to evaluate this. MATERIALS AND METHODS: Peripheral blood mononuclear cells were isolated from 90 healthy volunteers, divided into two aliquots, one irradiated with a 5 Gy dose and the other sham-treated (0 Gy), and assessed for damage-induced apoptosis after 24 hours. To investigate reproducibility, 10 individuals spanning the entire radiation-induced apoptotic range were tested three times each, with 3-6 months between replicates. RESULTS: We observed surprising heterogeneity in apoptosis among individuals, ranging from 21-62%. Biological replicates from a single individual, however, were completely concordant, suggesting the variability observed across individuals is not the result of stochastic or short-term effects. We found significantly higher radiation-induced apoptosis in males than in females (Mean: 41.0% vs. 30.7%; p < 3.5 × 10(-7)). Moreover, advancing age was associated with decreasing radiation-induced apoptosis in males (p = 0.01) but not females (p = 0.82). CONCLUSIONS: Our results provide evidence that the function of cellular pathways crucial for stress-induced apoptosis varies by sex and could decline with age in humans.
- Published
- 2014
- Full Text
- View/download PDF
34. Shared and independent colorectal cancer risk alleles in TGFβ-related genes in African and European Americans
- Author
-
Andrew D. Skol, Robert S. Sandler, Nathan A. Ellis, Ellie Hong, Sonia S. Kupfer, Rick A. Kittles, Temitope O. Keku, and Anton E. Ludvik
- Subjects
Male ,Cancer Research ,Linkage disequilibrium ,Original Manuscript ,Single-nucleotide polymorphism ,Genome-wide association study ,Bone Morphogenetic Protein 4 ,Biology ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,White People ,Smad7 Protein ,Gene Frequency ,Antigens, CD ,Humans ,SNP ,Genetic Predisposition to Disease ,Allele frequency ,Alleles ,Adaptor Proteins, Signal Transducing ,Aged ,Genetic association ,Genetics ,Genetic heterogeneity ,General Medicine ,Middle Aged ,Cadherins ,Black or African American ,Case-Control Studies ,Intercellular Signaling Peptides and Proteins ,Female ,Colorectal Neoplasms ,Imputation (genetics) ,Genome-Wide Association Study - Abstract
Genome-wide association studies (GWAS) in colorectal cancer (CRC) identified five regions near transforming growth factor β-related genes BMP4, GREM1, CDH1, SMAD7 and RPHN2. The true risk alleles remain to be identified in these regions, and their role in CRC risk in non-European populations has been understudied. Our previous work noted significant genetic heterogeneity between African Americans (AAs) and European Americans (EAs) for single nucleotide polymorphisms (SNPs) identified in GWAS. We hypothesized that associations may not have been replicated in AAs due to differential or independent genetic structures. In order to test this hypothesis, we genotyped 195 tagging SNPs across these five gene regions in 1194 CRC cases (795 AAs and 399 EAs) and 1352 controls (985 AAs and 367 EAs). Imputation was performed, and association testing of genotyped and imputed SNPs included ancestry, age and sex as covariates. In two of the five genes originally associated with CRC, we found evidence for association in AAs including rs1862748 in CDH1 (OR(Add) = 0.82, P = 0.02) and in GREM1 the SNPs rs10318 (OR(Rec) = 60.1, P = 0.01), rs11632715 (OR(Rec) = 2.36; P = 0.004) and rs12902616 (OR(Rec) = 1.28, P = 0.005), the latter which is in linkage disequilibrium with the previously identified SNP rs4779584. Testing more broadly for associations in these gene regions in AAs, we noted three statistically significant association peaks in GREM1 and RHPN2 that were not identified in EAs. We conclude that some CRC risk alleles are shared between EAs and AAs and others are population specific.
- Published
- 2014
- Full Text
- View/download PDF
35. Evaluation of Genetic Predisposition for MYCN-Amplified Neuroblastoma
- Author
-
Susan L. Cohn, Barbara E. Stranger, Lee D. McDaniel, Eric A. Hungate, Sharon J. Diskin, Andrew D. Skol, John M. Maris, Maura Diamond, Mark A. Applebaum, Zalman Vaksman, Samuel L. Volchenboum, and Kenan Onel
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Neuroblastoma ,0302 clinical medicine ,Gene Frequency ,Internal medicine ,Genetic variation ,medicine ,Genetic predisposition ,SNP ,Humans ,Genetic Predisposition to Disease ,neoplasms ,Genetic association ,N-Myc Proto-Oncogene Protein ,Gene Amplification ,Odds ratio ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Cancer research ,Brief Communications ,Genome-Wide Association Study - Abstract
To investigate genetic predispositions for MYCN-amplified neuroblastoma, we performed a meta-analysis of three genome-wide association studies totaling 615 MYCN-amplified high-risk neuroblastoma cases and 1869 MYCN-nonamplified non-high-risk neuroblastoma cases as controls using a fixed-effects model with inverse variance weighting. All statistical tests were two-sided. We identified a novel locus at 3p21.31 indexed by the single nucleotide polymorphism (SNP) rs80059929 (odds ratio [OR] = 2.95, 95% confidence interval [CI] = 2.17 to 4.02, Pmeta = 6.47 × 10-12) associated with MYCN-amplified neuroblastoma, which was replicated in 127 MYCN-amplified cases and 254 non-high-risk controls (OR = 2.30, 95% CI = 1.12 to 4.69, Preplication = .02). To confirm this signal is exclusive to MYCN-amplified tumors, we performed a second meta-analysis comparing 728 MYCN-nonamplified high-risk patients to identical controls. rs80059929 was not statistically significant in MYCN-nonamplified high-risk patients (OR = 1.24, 95% CI = 0.90 to 1.71, Pmeta = .19). SNP rs80059929 is within intron 16 in the KIF15 gene. Additionally, the previously reported LMO1 neuroblastoma risk locus was statistically significant only in patients with MYCN-nonamplified high-risk tumors (OR = 0.63, 95% CI = 0.53 to 0.75, Pmeta = 1.51 × 10-8; Pmeta = .95). Our results indicate that common genetic variation predisposes to different neuroblastoma genotypes, including the likelihood of somatic MYCN-amplification.
- Published
- 2016
36. The genetics of breast cancer risk in the post-genome era: thoughts on study design to move past BRCA and towards clinical relevance
- Author
-
Andrew D. Skol, Kenan Onel, and Mark M. Sasaki
- Subjects
0301 basic medicine ,Genome-wide association study ,Genetic Linkage ,Genes, BRCA2 ,Genes, BRCA1 ,Predisposition ,Penetrance ,Breast Neoplasms ,Genomics ,Review ,Environment ,Bioinformatics ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Missing heritability problem ,Genetics ,medicine ,Humans ,Family ,Genetic Predisposition to Disease ,skin and connective tissue diseases ,Genetic Association Studies ,Genetic association ,business.industry ,Clinical study design ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Next-generation sequencing ,Female ,Gene-Environment Interaction ,Breast Cancer Genetics ,business ,Linkage analysis - Abstract
More than 12 % of women will be diagnosed with breast cancer in their lifetime. Although there have been tremendous advances in elucidating genetic risk factors underlying both familial and sporadic breast cancer, much of the genetic contribution to breast cancer etiology remains unknown. The discovery of BRCA1 and BRCA2 over 20 years ago remains the seminal event in the field and has paved the way for the discovery of other high-penetrance susceptibility genes by linkage analysis. The advent of genome-wide association studies made possible the next wave of discoveries, in which over 80 low-penetrance and moderate-penetrance variants were identified. Although these studies were highly successful at discovering variants associated with both familial and sporadic breast cancer, the variants identified to date explain only 50 % of the heritability of breast cancer. In this review, we look back at the investigative strategies that have led to our current understanding of breast cancer genetics, consider the challenges of performing association studies in heterogeneous complex diseases such as breast cancer, and look ahead toward the types of study designs that may lead to the identification of the genetic variation accounting for the remaining missing heritability.
- Published
- 2016
- Full Text
- View/download PDF
37. A variant at 9p21.3 functionally implicates CDKN2B in paediatric B-cell precursor acute lymphoblastic leukaemia aetiology
- Author
-
Imge Hulur, Laurent Orsi, Eric R. Gamazon, Jérémie Rudant, Mignon L. Loh, Eva Ellinghaus, Eric A. Hungate, Ching-Hon Pui, Martin Stanulla, Andrew D. Skol, Timothy Best, Sapana Vora, Takaya Moriyama, Elizabeth Milne, Younghee Lee, Tiffany-Jane Evans, Jacqueline Clavel, Rodney J. Scott, Nancy J. Cox, Kenan Onel, Jun J. Yang, and Other departments
- Subjects
Male ,0301 basic medicine ,General Physics and Astronomy ,Genome-wide association study ,Exon ,CDKN2A ,hemic and lymphatic diseases ,CDKN2B ,CEBPB ,2.1 Biological and endogenous factors ,Aetiology ,Child ,Cancer ,African Americans ,Pediatric ,Genetics ,Multidisciplinary ,Chromosome Mapping ,Single Nucleotide ,Hispanic or Latino ,Splicing regulatory element ,3. Good health ,Child, Preschool ,Female ,Hispanic Americans ,Chromosomes, Human, Pair 9 ,Human ,Pair 9 ,Pediatric Cancer ,Science ,European Continental Ancestry Group ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Article ,Chromosomes ,White People ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Rare Diseases ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Humans ,Genetic Predisposition to Disease ,Polymorphism ,Preschool ,Cyclin-Dependent Kinase Inhibitor p15 ,Whites ,Human Genome ,Infant ,Genetic Variation ,General Chemistry ,Black or African American ,030104 developmental biology ,Case-Control Studies ,Genome-Wide Association Study - Abstract
Paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) is the most common cancer of childhood, yet little is known about BCP-ALL predisposition. In this study, in 2,187 cases of European ancestry and 5,543 controls, we discover and replicate a locus indexed by rs77728904 at 9p21.3 associated with BCP-ALL susceptibility (Pcombined=3.32 × 10−15, OR=1.72) and independent from rs3731217, the previously reported ALL-associated variant in this region. Of correlated SNPs tagged by this locus, only rs662463 is significant in African Americans, suggesting it is a plausible causative variant. Functional analysis shows that rs662463 is a cis-eQTL for CDKN2B, with the risk allele associated with lower expression, and suggests that rs662463 influences BCP-ALL risk by regulating CDKN2B expression through CEBPB signalling. Functional analysis of rs3731217 suggests it is associated with BCP-ALL by acting within a splicing regulatory element determining CDKN2A exon 3 usage (P=0.01). These findings provide new insights into the critical role of the CDKN2 locus in BCP-ALL aetiology., A risk variant located at 9p21.3 is associated with cancer risk in pediatric B-cell precursor acute lymphoblastic leukaemia. Here, the authors show that this variant affects the gene expression of the tumour suppressor gene Cdkn2b.
- Published
- 2016
- Full Text
- View/download PDF
38. A genome-wide meta-analysis of nodular sclerosing Hodgkin lymphoma identifies risk loci at 6p21.32
- Author
-
Dalin Li, Christopher K. Edlund, Pierre-Antoine Gourraud, Susan L. Slager, Victoria K. Cortessis, Louise C. Strong, Leslie L. Robison, Kenan Onel, James R. Cerhan, David Van Den Berg, Timothy Best, David V. Conti, Thomas M. Habermann, Andrew D. Skol, Zachary S. Fredericksen, Lawrence M. Weiss, Fredrick R. Schumacher, Brian K. Link, Wendy Cozen, Bharat N. Nathwani, Thomas M. Mack, Amie E. Hwang, Smita Bhatia, and Sally L. Glaser
- Subjects
Adult ,Male ,Adolescent ,Clinical Trials and Observations ,Immunology ,Genome-wide association study ,Single-nucleotide polymorphism ,Human leukocyte antigen ,Biology ,Polymorphism, Single Nucleotide ,Biochemistry ,Young Adult ,Risk Factors ,Polymorphism (computer science) ,Humans ,Genetic Predisposition to Disease ,Allele ,Child ,Genetics ,Haplotype ,Cell Biology ,Hematology ,Odds ratio ,Middle Aged ,Hodgkin Disease ,Haplotypes ,Case-Control Studies ,Child, Preschool ,Chromosomes, Human, Pair 6 ,Female ,Genome-Wide Association Study ,SNP array - Abstract
Nodular sclerosing Hodgkin lymphoma (NSHL) is a distinct, highly heritable Hodgkin lymphoma subtype. We undertook a genome-wide meta-analysis of 393 European-origin adolescent/young adult NSHL patients and 3315 controls using the Illumina Human610-Quad Beadchip and Affymetrix Genome-Wide Human SNP Array 6.0. We identified 3 single nucleotide polymorphisms (SNPs) on chromosome 6p21.32 that were significantly associated with NSHL risk: rs9268542 (P = 5.35 × 10−10), rs204999 (P = 1.44 × 10−9), and rs2858870 (P = 1.69 × 10−8). We also confirmed a previously reported association in the same region, rs6903608 (P = 3.52 × 10−10). rs204999 and rs2858870 were weakly correlated (r2 = 0.257), and the remaining pairs of SNPs were not correlated (r2 < 0.1). In an independent set of 113 NSHL cases and 214 controls, 2 SNPs were significantly associated with NSHL and a third showed a comparable odds ratio (OR). These SNPs are found on 2 haplotypes associated with NSHL risk (rs204999-rs9268528-rs9268542-rs6903608-rs2858870; AGGCT, OR = 1.7, P = 1.71 × 10−6; GAATC, OR = 0.4, P = 1.16 × 10−4). All individuals with the GAATC haplotype also carried the HLA class II DRB1*0701 allele. In a separate analysis, the DRB1*0701 allele was associated with a decreased risk of NSHL (OR = 0.5, 95% confidence interval = 0.4, 0.7). These data support the importance of the HLA class II region in NSHL etiology.
- Published
- 2012
- Full Text
- View/download PDF
39. Abstract 1625: Clinical biomarkers of survival in renal cell carcinoma patients treated with sorafenib
- Author
-
Andrew D. Skol, Nancy Klauber-DeMore, Kari Alitalo, Yuri K. Peterson, Federico Innocenti, Carol Peña, Amy S. Etheridge, Dylan M. Glubb, Daniel J. Crona, Veli-Matti Leppänen, and Eleanor Hilliard
- Subjects
Sorafenib ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Renal cell carcinoma ,Internal medicine ,Medicine ,business ,medicine.disease ,medicine.drug - Abstract
Purpose: To discover genetic determinants of overall survival (OS) in metastatic renal cell carcinoma (mRCC) patients treated with sorafenib. Patients and Methods: Information for 11,117 germline DNA variants from 56 genes was obtained, using both Illumina GoldenGate genotyping and imputation, from 295 patients originally enrolled in the phase III TARGET trial (155 sorafenib and 140 placebo patients). OS was this genotyping study's primary endpoint, and associations between variants and OS were tested using multivariate Cox regression. Associations were considered significant if P Results: Five intragenic variants significantly associated with OS. Among patients in the sorafenib arm, VEGFA rs1885657 (HR=17.3, 95% CI 5.7-52.7; P=1.4x10-4), ITGAV rs3816375 (HR=5.9, 95% CI 2.1-16.4; P=4.9x10-4), and WWOX rs8047917 (HR=4.1, 95% CI 1.9-8.8; P=3.3x10-4) associated with shorter OS. Among patients from both arms, FLT4 rs307826 (HR=13.8, 95% CI 3.0-62.6; P=1.2x10-4) and VEGFA rs3024987 (HR=3.0, 95% CI 1.7-5.4; P=8.3x10-5) associated with shorter OS. No significant associations were discovered in the placebo arm. Bioinformatic evidence was used to prioritize rs1885657, rs58159269 (in complete linkage disequilibrium with rs1885657), and rs307826. rs1885657 and rs58159269 increased luciferase activity in human endothelial (TIME, LPEC) and mRCC (Caki-1) cells, and rs58159269 increased cell proliferation/reduced sorafenib cytotoxicity, and reduced endothelial tube formation in TIME cells. rs307826 increased VEGFR-3 phosphorylation, and led to post-translational VEGFR-3 processing differences in HUVECs. Conclusions: Regulatory VEGFA and FLT4 variants associated with OS in mRCC patients induce cellular alterations that affect angiogenesis and sorafenib activity. These are novel and potentially impactful clinical biomarkers for mRCC and sorafenib, and their relevance to other multikinase inhibitors used in mRCC should be tested. Citation Format: Daniel James Crona, Andrew D. Skol, Veli-Matti Leppänen, Dylan M. Glubb, Amy S. Etheridge, Eleanor Hilliard, Carol Peña, Yuri K. Peterson, Nancy Klauber-DeMore, Kari K. Alitalo, Federico Innocenti. Clinical biomarkers of survival in renal cell carcinoma patients treated with sorafenib [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1625.
- Published
- 2018
- Full Text
- View/download PDF
40. Genome-Wide Association Studies and the Problem of Relatedness Among Advanced Intercross Lines and Other Highly Recombinant Populations
- Author
-
Andrew D. Skol, Jackie E. Lim, Kaitlin E. Samocha, Abraham A. Palmer, Greta Sokoloff, Mark Abney, and Riyan Cheng
- Subjects
Genetic Linkage ,Quantitative Trait Loci ,Locus (genetics) ,Genome-wide association study ,Investigations ,Biology ,Quantitative trait locus ,Polymorphism, Single Nucleotide ,Genome ,Mice ,Gene mapping ,Inbred strain ,Genetic linkage ,Genetics ,Animals ,Humans ,Crosses, Genetic ,Genetic association ,Recombination, Genetic ,Behavior, Animal ,Chromosome Mapping ,Computational Biology ,Chromosomes, Mammalian ,Phenotype ,Genome-Wide Association Study - Abstract
Model organisms offer many advantages for the genetic analysis of complex traits. However, identification of specific genes is often hampered by a lack of recombination between the genomes of inbred progenitors. Recently, genome-wide association studies (GWAS) in humans have offered gene-level mapping resolution that is possible because of the large number of accumulated recombinations among unrelated human subjects. To obtain analogous improvements in mapping resolution in mice, we used a 34th generation advanced intercross line (AIL) derived from two inbred strains (SM/J and LG/J). We used simulations to show that familial relationships among subjects must be accounted for when analyzing these data; we then used a mixed model that included polygenic effects to address this problem in our own analysis. Using a combination of F2 and AIL mice derived from the same inbred progenitors, we identified genome-wide significant, subcentimorgan loci that were associated with methamphetamine sensitivity, (e.g., chromosome 18; LOD = 10.5) and non-drug-induced locomotor activity (e.g., chromosome 8; LOD = 18.9). The 2-LOD support interval for the former locus contains no known genes while the latter contains only one gene (Csmd1). This approach is broadly applicable in terms of phenotypes and model organisms and allows GWAS to be performed in multigenerational crosses between and among inbred strains where familial relatedness is often unavoidable.
- Published
- 2010
- Full Text
- View/download PDF
41. Genome-wide association study to identify novel loci associated with therapy-related myeloid leukemia susceptibility
- Author
-
Mekhala Banerjee, Andrew D. Skol, Nancy J. Cox, Michelle M. Le Beau, Kenan Onel, Richard A. Walgren, Jeffrey Knight, Jin Shao, Richard A. Larson, Thelma R. Tennant, Timothy A. Graubert, Darcie Hastings, Abhijit Shinde, and James M. Allan
- Subjects
Adult ,Male ,Adolescent ,Quantitative Trait Loci ,Immunology ,Single-nucleotide polymorphism ,Genome-wide association study ,Polymorphism, Single Nucleotide ,Biochemistry ,Cohort Studies ,Young Adult ,hemic and lymphatic diseases ,Humans ,Medicine ,Genetic Predisposition to Disease ,Allele ,Aged ,Aged, 80 and over ,Myeloid Neoplasia ,business.industry ,Case-control study ,Myeloid leukemia ,Cancer ,Neoplasms, Second Primary ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Leukemia ,Leukemia, Myeloid ,Case-Control Studies ,Female ,business ,Genome-Wide Association Study ,Chronic myelogenous leukemia - Abstract
Therapy-related acute myeloid leukemia (t-AML) is a rare but fatal complication of cytotoxic therapy. Whereas sporadic cancer results from interactions between complex exposures and low-penetrance alleles, t-AML results from an acute exposure to a limited number of potent genotoxins. Consequently, we hypothesized that the effect sizes of variants associated with t-AML would be greater than in sporadic cancer, and, therefore, that these variants could be detected even in a modest-sized cohort. To test this, we undertook an association study in 80 cases and 150 controls using Affymetrix Mapping 10K arrays. Even at nominal significance thresholds, we found a significant excess of associations over chance; for example, although 6 associations were expected at P less than .001, we found 15 (Penrich = .002). To replicate our findings, we genotyped the 10 most significantly associated single nucleotide polymorphisms (SNPs) in an independent t-AML cohort (n = 70) and obtained evidence of association with t-AML for 3 SNPs in the subset of patients with loss of chromosomes 5 or 7 or both, acquired abnormalities associated with prior exposure to alkylator chemotherapy. Thus, we conclude that the effect of genetic factors contributing to cancer risk is potentiated and more readily discernable in t-AML compared with sporadic cancer.
- Published
- 2009
- Full Text
- View/download PDF
42. Interaction of Prenatal Exposure to Cigarettes and MAOA Genotype in Pathways to Youth Antisocial Behavior
- Author
-
Vanja Dukic, Kate E. Pickett, Andrew D. Skol, Nancy J. Cox, Gretchen Biesecker, Emily O. Kistner, Daniel S. Pine, Kristen Kasza, James L. Burns, Rosalind J. Wright, Edwin H. Cook, Robert James Blair, Lauren S. Wakschlag, and Bennett L. Leventhal
- Subjects
Adult ,Male ,Adolescent ,Genotype ,Monoamine oxidase ,Environment ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,0302 clinical medicine ,Pregnancy ,Risk Factors ,Genetic predisposition ,medicine ,developmental psychopathology ,Humans ,Genetic Predisposition to Disease ,MAOA ,Young adult ,Gene–environment interaction ,Sex Distribution ,Molecular Biology ,Monoamine Oxidase ,Genetics ,biology ,gene × environment interaction ,Smoking ,Social Behavior Disorders ,medicine.disease ,030227 psychiatry ,3. Good health ,prenatal smoking ,Psychiatry and Mental health ,Phenotype ,Conduct disorder ,Adolescent Behavior ,Prenatal Exposure Delayed Effects ,biology.protein ,Female ,Monoamine oxidase A ,Psychology ,030217 neurology & neurosurgery ,Developmental psychopathology ,Follow-Up Studies - Abstract
Genetic susceptibility to antisocial behavior may increase fetal sensitivity to prenatal exposure to cigarette smoke. Testing putative gene x exposure mechanisms requires precise measurement of exposure and outcomes. We tested whether a functional polymorphism in the gene encoding the enzyme monoamine oxidase A (MAOA) interacts with exposure to predict pathways to adolescent antisocial behavior. We assessed both clinical and information-processing outcomes. One hundred seventy-six adolescents and their mothers participated in a follow-up of a pregnancy cohort with well-characterized exposure. A sex-specific pattern of gene x exposure interaction was detected. Exposed boys with the low-activity MAOA 5' uVNTR (untranslated region variable number of tandem repeats) genotype were at increased risk for conduct disorder (CD) symptoms. In contrast, exposed girls with the high-activity MAOA uVNTR genotype were at increased risk for both CD symptoms and hostile attribution bias on a face-processing task. There was no evidence of a gene-environment correlation (rGE). Findings suggest that the MAOA uVNTR genotype, prenatal exposure to cigarettes and sex interact to predict antisocial behavior and related information-processing patterns. Future research to replicate and extend these findings should focus on elucidating how gene x exposure interactions may shape behavior through associated changes in brain function.
- Published
- 2009
43. Screening of 134 Single Nucleotide Polymorphisms (SNPs) Previously Associated With Type 2 Diabetes Replicates Association With 12 SNPs in Nine Genes
- Author
-
Richard N. Bergman, Francis S. Collins, Kimberly F. Doheny, Cristen J. Willer, Richard M. Watanabe, Lori L. Bonnycastle, Eboni Smith, Andrew D. Skol, Timo T. Valle, Sareena T. Enloe, John Petrie, Andrew G. Sprau, Karen N. Conneely, Thomas A. Buchanan, Jaakko Tuomilehto, Michael R. Erdos, Elizabeth W. Pugh, Karen L. Mohlke, William L. Duren, Michael Boehnke, Peter S. Chines, Amy J. Swift, Narisu Narisu, Maurine Tong, Laura J. Scott, Heather M. Stringham, and Anne U. Jackson
- Subjects
Blood Glucose ,Male ,Linkage disequilibrium ,Endocrinology, Diabetes and Metabolism ,Population ,030209 endocrinology & metabolism ,Single-nucleotide polymorphism ,Type 2 diabetes ,Biology ,Polymorphism, Single Nucleotide ,Body Mass Index ,03 medical and health sciences ,0302 clinical medicine ,Polymorphism (computer science) ,Internal Medicine ,medicine ,Humans ,Insulin ,SNP ,Genetic Testing ,education ,Aged ,030304 developmental biology ,Genetics ,0303 health sciences ,education.field_of_study ,Chromosome Mapping ,Infant ,Fasting ,Middle Aged ,Tag SNP ,medicine.disease ,HNF1A ,Diabetes Mellitus, Type 2 ,Female - Abstract
More than 120 published reports have described associations between single nucleotide polymorphisms (SNPs) and type 2 diabetes. However, multiple studies of the same variant have often been discordant. From a literature search, we identified previously reported type 2 diabetes–associated SNPs. We initially genotyped 134 SNPs on 786 index case subjects from type 2 diabetes families and 617 control subjects with normal glucose tolerance from Finland and excluded from analysis 20 SNPs in strong linkage disequilibrium (r2 > 0.8) with another typed SNP. Of the 114 SNPs examined, we followed up the 20 most significant SNPs (P < 0.10) on an additional 384 case subjects and 366 control subjects from a population-based study in Finland. In the combined data, we replicated association (P < 0.05) for 12 SNPs: PPARG Pro12Ala and His447, KCNJ11 Glu23Lys and rs5210, TNF −857, SLC2A2 Ile110Thr, HNF1A/TCF1 rs2701175 and GE117881_360, PCK1 −232, NEUROD1 Thr45Ala, IL6 −598, and ENPP1 Lys121Gln. The replication of 12 SNPs of 114 tested was significantly greater than expected by chance under the null hypothesis of no association (P = 0.012). We observed that SNPs from genes that had three or more previous reports of association were significantly more likely to be replicated in our sample (P = 0.03), although we also replicated 4 of 58 SNPs from genes that had only one previous report of association.
- Published
- 2007
- Full Text
- View/download PDF
44. Whole-exome Sequence Analysis Implicates Rare Il17REL Variants in Familial and Sporadic Inflammatory Bowel Disease
- Author
-
Inga Peter, Dermot P.B. McGovern, Lei Huang, Steven R. Brant, Stacy A. Kahn, Riyue Bao, Barbara S. Kirschner, Judy H. Cho, Eric A. Hungate, Kenan Onel, Andrew D. Skol, Mark S. Silverberg, Mark M. Sasaki, and James M. Allan
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,Population ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Young Adult ,Obligate carrier ,medicine ,Immunology and Allergy ,Humans ,Exome ,Genetic Predisposition to Disease ,Family history ,education ,Exome sequencing ,Genetic testing ,Aged ,Genetics ,education.field_of_study ,Receptors, Interleukin-17 ,medicine.diagnostic_test ,business.industry ,Gastroenterology ,High-Throughput Nucleotide Sequencing ,Odds ratio ,Middle Aged ,Inflammatory Bowel Diseases ,Prognosis ,Pedigree ,Minor allele frequency ,030104 developmental biology ,Phenotype ,Case-Control Studies ,Female ,business - Abstract
BACKGROUND Rare variants (
- Published
- 2015
45. Integrated genomic analysis suggests MLL3 is a novel candidate susceptibility gene for familial nasopharyngeal carcinoma
- Author
-
Ezra E.W. Cohen, Rachelle Lorenz Chambers, Kenan Onel, Everett E. Vokes, Lindsay Rhodes, Mark M. Sasaki, Riyue Bao, and Andrew D. Skol
- Subjects
Epidemiology ,Biology ,medicine.disease_cause ,Medical and Health Sciences ,Article ,Germline ,Germline mutation ,Clinical Research ,otorhinolaryngologic diseases ,medicine ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Genetic Testing ,Dental/Oral and Craniofacial Disease ,Aetiology ,Gene ,Exome ,Exome sequencing ,Germ-Line Mutation ,Cancer ,Mutation ,Nasopharyngeal Carcinoma ,Carcinoma ,Human Genome ,Nasopharyngeal Neoplasms ,Genomics ,medicine.disease ,Stop codon ,DNA-Binding Proteins ,stomatognathic diseases ,Oncology ,Nasopharyngeal carcinoma ,Biotechnology - Abstract
Background: Little is known about genetic factors associated with nasopharyngeal carcinoma (NPC). To gain insight into NPC etiology, we performed whole exome sequencing on germline and tumor DNA from three closely related family members with NPC. Methods: The family was ascertained through the Pediatric Familial Cancer Clinic at The University of Chicago (Chicago, IL). The diagnosis of NPC was confirmed pathologically for each individual. For each sample sequenced, 97.3% of the exome was covered at 5×, with an average depth of 44×. Candidate germline and somatic variants associated with NPC were identified and prioritized using a custom pipeline. Results: We discovered 72 rare deleterious germline variants in 56 genes shared by all three individuals. Of these, only three are in previously identified NPC-associated genes, all of which are located within MLL3, a gene known to be somatically altered in NPC. One variant introduces an early stop codon in MLL3, which predicts complete loss-of-function. Tumor DNA analysis revealed somatic mutations and Epstein–Barr virus (EBV) integration events; none, however, were shared among all three individuals. Conclusions: These data suggest that inherited mutations in MLL3 may have predisposed these three individuals from a single family to develop NPC, and may cooperate with individually acquired somatic mutations or EBV integration events in NPC etiology. Impact: Our finding is the first instance of a plausible candidate high penetrance inherited mutation predisposing to NPC. Cancer Epidemiol Biomarkers Prev; 24(8); 1222–8. ©2015 AACR.
- Published
- 2015
46. Whole Exome Sequencing Elucidates Genomic Evolution of Extramedullary Acute Myeloid Leukemia (EM-AML) from Bone Marrow Acute Myeloid Leukemia (BM-AML)
- Author
-
Friedrich Stölzel, Andrew D. Skol, Kai Lee Yap, Kazuma Kiyotani, Yusuke Nakamura, Friederike Kuithan, Gordana Raca, and Kenan Onel
- Subjects
Cancer Research ,medicine.anatomical_structure ,Genetics ,medicine ,Cancer research ,Myeloid leukemia ,Bone marrow ,Biology ,Molecular Biology ,Exome sequencing - Published
- 2016
- Full Text
- View/download PDF
47. Modest evidence for linkage and possible confirmation of association between NOTCH4 and schizophrenia in a large veterans affairs cooperative study sample
- Author
-
Chang En Yu, Felicitas Mena, Stephen F. Bingham, T. Keith, Paul Rundell, Debby Tsuang, Michael Boehnke, Joseph F. Collins, Andrew D. Skol, Ming T. Tsuang, David G. Weiss, Sarita Prabhudesai, Gerard D. Schellenberg, S.L. Haverstock, John R. Pepple, Clinton T. Baldwin, A.S. Menon, Stephen V. Faraone, Keith A. Young, and F. Sauter
- Subjects
Genetic Markers ,Male ,Linkage disequilibrium ,Genetic Linkage ,Receptors, Cell Surface ,Schizoaffective disorder ,Biology ,Linkage Disequilibrium ,Genetic determinism ,Gene Frequency ,Genetic linkage ,Proto-Oncogene Proteins ,medicine ,Humans ,Allele ,Receptor, Notch4 ,Veterans Affairs ,Allele frequency ,Alleles ,Genetics (clinical) ,Family Health ,Genetics ,Receptors, Notch ,Haplotype ,medicine.disease ,Haplotypes ,Schizophrenia ,Female ,Lod Score - Abstract
Wei and Hemmings [2000: Nat Genet 25:376-377], using 80 British parent-offspring trios, identified a number of NOTCH4 variants and haplotypes that showed statistically significant evidence of association to schizophrenia. Specifically, the 10 repeat allele of a (CTG)(n) marker and the 8 repeat allele of a (TAA)(n) marker demonstrated excess transmission to affected individuals; SNP21 and haplotypes SNP2-(CTG)(n) and SNP12-SNP2-(CTG)(n) also showed significant associations. In an attempt to replicate these findings, we tested for linkage and association between the same five markers used by Wei and Hemmings in 166 families collected from a multi-center study conducted by the Department of Veterans Affairs (DVA) Cooperative Study Program (CSP). The families include 392 affected subjects (schizophrenia or schizoaffective disorder, depressed) and 216 affected sibling pairs. The families represent a mix of European Americans (n = 62, 37%), African Americans (n = 60, 36%), and racially mixed or other races (n = 44, 27%). We identified moderate evidence for linkage in the pooled race sample (LOD = 1.25) and found excess transmission of the 8 (P = 0.06) and 13 (P = 0.04) repeat alleles of the (TAA)(n) marker to African American schizophrenic subjects. The 8 and 13 repeat alleles were previously identified to be positively associated with schizophrenia by Wei and Hemmings [2000: Nat Genet 25:376-377] and Sklar et al. [2001: Nat Genet 28:126-128], respectively.
- Published
- 2003
- Full Text
- View/download PDF
48. Probability of Detection of Genotyping Errors and Mutations as Inheritance Inconsistencies in Nuclear-Family Data
- Author
-
Julie A. Douglas, Andrew D. Skol, and Michael Boehnke
- Subjects
Male ,Genotype ,Genetic Linkage ,Biology ,Polymorphism, Single Nucleotide ,Sensitivity and Specificity ,Statistical power ,Nuclear Family ,03 medical and health sciences ,symbols.namesake ,Gene Frequency ,Resampling ,Genetics ,Humans ,Computer Simulation ,Genetics(clinical) ,Allele ,Genotyping ,Nuclear family ,Alleles ,Genetics (clinical) ,Probability ,030304 developmental biology ,0303 health sciences ,Models, Genetic ,030305 genetics & heredity ,Inheritance (genetic algorithm) ,Chromosome Mapping ,Articles ,Pedigree ,Research Design ,Mutation ,Mutation (genetic algorithm) ,Mendelian inheritance ,symbols ,Female - Abstract
Gene-mapping studies routinely rely on checking for Mendelian transmission of marker alleles in a pedigree, as a means of screening for genotyping errors and mutations, with the implicit assumption that, if a pedigree is consistent with Mendel's laws of inheritance, then there are no genotyping errors. However, the occurrence of inheritance inconsistencies alone is an inadequate measure of the number of genotyping errors, since the rate of occurrence depends on the number and relationships of genotyped pedigree members, the type of errors, and the distribution of marker-allele frequencies. In this article, we calculate the expected probability of detection of a genotyping error or mutation as an inheritance inconsistency in nuclear-family data, as a function of both the number of genotyped parents and offspring and the marker-allele frequency distribution. Through computer simulation, we explore the sensitivity of our analytic calculations to the underlying error model. Under a random-allele-error model, we find that detection rates are 51%-77% for multiallelic markers and 13%-75% for biallelic markers; detection rates are generally lower when the error occurs in a parent than in an offspring, unless a large number of offspring are genotyped. Errors are especially difficult to detect for biallelic markers with equally frequent alleles, even when both parents are genotyped; in this case, the maximum detection rate is 34% for four-person nuclear families. Error detection in families in which parents are not genotyped is limited, even with multiallelic markers. Given these results, we recommend that additional error checking (e.g., on the basis of multipoint analysis) be performed, beyond routine checking for Mendelian consistency. Furthermore, our results permit assessment of the plausibility of an observed number of inheritance inconsistencies for a family, allowing the detection of likely pedigree-rather than genotyping-errors in the early stages of a genome scan. Such early assessments are valuable in either the targeting of families for resampling or discontinued genotyping.
- Published
- 2002
- Full Text
- View/download PDF
49. Integrative genetic analysis suggests that skin color modifies the genetic architecture of melanoma
- Author
-
Andrew D. Skol, Kenan Onel, Eric R. Gamazon, Nancy J. Cox, and Imge Hulur
- Subjects
Melanomas ,0301 basic medicine ,Heredity ,Skin Neoplasms ,lcsh:Medicine ,Skin Pigmentation ,Genome-wide association study ,Geographical Locations ,Gene Frequency ,Medicine and Health Sciences ,lcsh:Science ,Melanoma ,Genetics ,Multidisciplinary ,medicine.diagnostic_test ,Genomics ,Europe ,Genetic Mapping ,Oncology ,Research Article ,Variant Genotypes ,Biology ,Genetic Predisposition ,Polymorphism, Single Nucleotide ,White People ,03 medical and health sciences ,Genome-Wide Association Studies ,medicine ,Genetic predisposition ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,neoplasms ,Genetic testing ,Models, Genetic ,lcsh:R ,Cancers and Neoplasms ,Biology and Life Sciences ,Computational Biology ,Genetic Variation ,Human Genetics ,Genome Analysis ,medicine.disease ,Genetic architecture ,030104 developmental biology ,Genetic Loci ,Genetics of Disease ,People and Places ,Expression quantitative trait loci ,Cutaneous melanoma ,lcsh:Q ,Skin cancer ,Genome-Wide Association Study - Abstract
Melanoma is the deadliest form of skin cancer and presents a significant health care burden in many countries. In addition to ultraviolet radiation in sunlight, the main causal factor for melanoma, genetic factors also play an important role in melanoma susceptibility. Although genome-wide association studies have identified many single nucleotide polymorphisms associated with melanoma, little is known about the proportion of disease risk attributable to these loci and their distribution throughout the genome. Here, we investigated the genetic architecture of melanoma in 1,888 cases and 990 controls of European non-Hispanic ancestry. We estimated the overall narrow-sense heritability of melanoma to be 0.18 (P < 0.03), indicating that genetics contributes significantly to the risk of sporadically-occurring melanoma. We then demonstrated that only a small proportion of this risk is attributable to known risk variants, suggesting that much remains unknown of the role of genetics in melanoma. To investigate further the genetic architecture of melanoma, we partitioned the heritability by chromosome, minor allele frequency, and functional annotations. We showed that common genetic variation contributes significantly to melanoma risk, with a risk model defined by a handful of genomic regions rather than many risk loci distributed throughout the genome. We also demonstrated that variants affecting gene expression in skin account for a significant proportion of the heritability, and are enriched among melanoma risk loci. Finally, by incorporating skin color into our analyses, we observed both a shift in significance for melanoma-associated loci and an enrichment of expression quantitative trait loci among melanoma susceptibility variants. These findings suggest that skin color may be an important modifier of melanoma risk. We speculate that incorporating skin color and other non-genetic factors into genetic studies may allow for an improved understanding of melanoma susceptibility and guide future investigations to identify melanoma risk genes.
- Published
- 2017
- Full Text
- View/download PDF
50. Examination of genetic linkage of chromosome 15 to schizophrenia in a large Veterans Affairs Cooperative Study sample
- Author
-
S.L. Haverstock, Felicitas Mena, Debby Tsuang, Michael Boehnke, John R. Pepple, Andrew D. Skol, A. Shrikumar Menon, Gerard D. Schellenberg, Sarita Prabhudesai, Fred Sauter, Stephen F. Bingham, Keith A. Young, Charlene Baldwin, Joseph F. Collins, Darren Bisset, David G. Weiss, Ming T. Tsuang, and Stephen V. Faraone
- Subjects
Genetics ,Linkage (software) ,CHRNA7 ,Biology ,medicine.disease ,Chromosome 15 ,Schizophrenia ,Genetic linkage ,biology.protein ,medicine ,Sibling ,Veterans Affairs ,Genetics (clinical) ,Diagnosis of schizophrenia - Abstract
Previous studies have reported genetic linkage evidence for a schizophrenia gene on chromosome 15q. Here, chromosome 15 was examined by genetic linkage analysis using 166 schizophrenia families, each with two or more affected subjects. The families, assembled from multiple centers by the Department of Veterans Affairs Cooperative Study Program, consisted of 392 sampled affected subjects and 216 affected sibling pairs. By DSM-III-R criteria, 360 subjects (91.8%) had a diagnosis of schizophrenia and 32 (8.2%) were classified as schizo-affective disorder, depressed. Participating families had diverse ethnic backgrounds. The largest single group were northern European American families (n = 62, 37%), but a substantial proportion was African American kindreds (n = 60, 36%). The chromosome 15 markers tested were spaced at intervals of approximately 10 cM over the entire chromosome and 2-5 cM for the region surrounding the alpha-7 nicotinic cholinergic receptor subunit gene (CHRNA7). These markers were genotyped and the data analyzed using semiparametric affecteds-only linkage analysis. In the European American families, there was a maximum Z-score of 1.65 between markers D15S165 and D15S1010. These markers are within 1 cM from CHRNA-7, the site previously implicated in schizophrenia. However, there was no evidence for linkage to this region in the African America kindreds.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.