Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Br J Haematol ; 200(4): 489-493, 2023 02.
Article in English | MEDLINE | ID: mdl-36349721

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.


Subject(s)
Neoplasms, Second Primary , Neoplasms , Humans , Germ-Line Mutation , Neoplasms/genetics , Mutation , Genetic Predisposition to Disease , Genetic Testing , Neoplasms, Second Primary/genetics
2.
Cell Rep Med ; 3(6): 100644, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35617957

ABSTRACT

Over the last decade, sequencing of primary tumors has clarified the genetic underpinnings of Wilms tumor but has not affected therapy, outcome, or toxicity. We now sharpen our focus on relapse samples from the umbrella AREN03B2 study. We show that over 40% of relapse samples contain mutations in SIX1 or genes of the MYCN network, drivers of progenitor proliferation. Not previously seen in large studies of primary Wilms tumors, DIS3 and TERT are now identified as recurrently mutated. The analysis of primary-relapse tumor pairs suggests that 11p15 loss of heterozygosity (and other copy number changes) and mutations in WT1 and MLLT1 typically occur early, but mutations in SIX1, MYCN, and WTX are late developments in some individuals. Most strikingly, 75% of relapse samples had gain of 1q, providing strong conceptual support for studying circulating tumor DNA in clinical trials to better detect 1q gain earlier and monitor response.


Subject(s)
Kidney Neoplasms , Wilms Tumor , Child , Genes, Wilms Tumor , Homeodomain Proteins/genetics , Humans , Kidney Neoplasms/genetics , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Recurrence, Local/genetics , Wilms Tumor/genetics
3.
Pharmacogenomics J ; 22(1): 82-88, 2022 02.
Article in English | MEDLINE | ID: mdl-34775477

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).


Subject(s)
Hypertension/chemically induced , Hypertension/genetics , Phosphatidylinositol 3-Kinase/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aged , Bevacizumab/adverse effects , Biomarkers , Double-Blind Method , Female , Genetic Testing , Humans , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide , Predictive Value of Tests , Risk Assessment , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
5.
Cancer Prev Res (Phila) ; 14(4): 441-454, 2021 04.
Article in English | MEDLINE | ID: mdl-33419763

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.


Subject(s)
Biomarkers, Tumor/genetics , Exome , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Germ-Line Mutation , Neoplastic Syndromes, Hereditary/pathology , Receptor, ErbB-2/genetics , Adolescent , Adult , Aged , Child , DNA Mutational Analysis , Female , Genetic Testing , Humans , Male , Neoplastic Syndromes, Hereditary/genetics , Pedigree , Exome Sequencing , Young Adult
6.
Elife ; 92020 11 09.
Article in English | MEDLINE | ID: mdl-33164750

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.


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.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetic Retinopathy/genetics , Gene Expression Profiling , Glucose/toxicity , Lymphocytes/drug effects , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics , Transcriptome , Tumor Suppressor Proteins/genetics , Adult , Case-Control Studies , Cell Line, Transformed , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/metabolism , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Lymphocytes/metabolism , Male , Mendelian Randomization Analysis , Proto-Oncogene Proteins/metabolism , Quantitative Trait Loci , Tumor Suppressor Proteins/metabolism , Young Adult
7.
Science ; 369(6509)2020 09 11.
Article in English | MEDLINE | ID: mdl-32913072

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.


Subject(s)
Gene Expression Regulation , Gene Expression , Sex Characteristics , Chromosomes, Human, X/genetics , Disease/genetics , Epigenesis, Genetic , Female , Genetic Variation , Genome-Wide Association Study , Humans , Male , Organ Specificity , Promoter Regions, Genetic , Quantitative Trait Loci , Sex Factors
8.
Cancer Res ; 79(1): 231-241, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30385613

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.


Subject(s)
Carcinoma, Renal Cell/mortality , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms/mortality , Mutation , Sorafenib/therapeutic use , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Cell Proliferation , Double-Blind Method , Female , Follow-Up Studies , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Prognosis , Survival Rate , Tumor Cells, Cultured , Young Adult
9.
J Natl Cancer Inst ; 109(10)2017 10 01.
Article in English | MEDLINE | ID: mdl-29117357

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.


Subject(s)
Gene Amplification , Genetic Predisposition to Disease , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Case-Control Studies , Gene Expression Regulation, Neoplastic , Gene Frequency , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
10.
Cancer Manag Res ; 9: 397-410, 2017.
Article in English | MEDLINE | ID: mdl-28979163

ABSTRACT

Gene signatures have been associated with outcome in pediatric acute lymphoblastic leukemia (ALL) and other malignancies. However, determining the molecular drivers of these expression changes remains challenging. In ALL blasts, the p53 tumor suppressor is the primary regulator of the apoptotic response to genotoxic chemotherapy, which is predictive of outcome. Consequently, we hypothesized that the normal p53-regulated apoptotic response to DNA damage would be altered in ALL and that this alteration would influence drug response and treatment outcome. To test this, we first used global expression profiling in related human B-lineage lymphoblastoid cell lines with either wild type or mutant TP53 to characterize the normal p53-mediated transcriptional response to ionizing radiation (IR) and identified 747 p53-regulated apoptotic target genes. We then sorted these genes into six temporal expression clusters (TECs) based upon differences over time in their IR-induced p53-regulated gene expression patterns, and found that one cluster (TEC1) was associated with multidrug resistance in leukemic blasts in one cohort of children with ALL and was an independent predictor of survival in two others. Therefore, by investigating p53-mediated apoptosis in vitro, we identified a gene signature significantly associated with drug resistance and treatment outcome in ALL. These results suggest that intersecting pathway-derived and clinically derived expression data may be a powerful method to discover driver gene signatures with functional and clinical implications in pediatric ALL and perhaps other cancers as well.

11.
PLoS One ; 12(10): e0185730, 2017.
Article in English | MEDLINE | ID: mdl-28973033

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.


Subject(s)
Gene Frequency , Genetic Loci , Genetic Predisposition to Disease , Melanoma/genetics , Skin Neoplasms/genetics , Skin Pigmentation/genetics , Genetic Testing , Genetic Variation , Genome-Wide Association Study , Humans , Models, Genetic , Polymorphism, Single Nucleotide , White People/genetics
12.
Neurotoxicol Teratol ; 61: 82-91, 2017 05.
Article in English | MEDLINE | ID: mdl-28163169

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 (ß=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 (ß=0.813, p=0.022). Boys with MAOA-H had more DB as a function of PSE, controlling for PTE (ß=0.774, p=0.015), and as a function of PTE, controlling for PSE (ß=0.362, p=0.037). Boys with MAOA-L did not show this susceptibility. MAOA did not interact with PSE (ß=-0.133, p=0.561) nor PTE (ß=-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.


Subject(s)
Monoamine Oxidase/adverse effects , Prenatal Exposure Delayed Effects/genetics , Problem Behavior , Stress, Psychological/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Male , Pregnancy , Prospective Studies , Sex Characteristics , Tobacco Smoking/adverse effects , Young Adult
13.
Breast Cancer Res ; 18(1): 99, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27716388

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.


Subject(s)
Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Genetic Predisposition to Disease , Genomics , Environment , Family , Female , Gene-Environment Interaction , Genes, BRCA1 , Genes, BRCA2 , Genetic Association Studies , Genetic Linkage , Genetic Variation , Genome-Wide Association Study , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Penetrance , Risk Assessment
14.
Nat Commun ; 7: 10635, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26868379

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Black or African American/genetics , Case-Control Studies , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 9/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Hispanic or Latino/genetics , Humans , Infant , Male , Polymorphism, Single Nucleotide , White People/genetics
15.
Inflamm Bowel Dis ; 22(1): 20-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26480299

ABSTRACT

BACKGROUND: Rare variants (<1%) likely contribute significantly to risk for common diseases such as inflammatory bowel disease (IBD) in specific patient subsets, such as those with high familiality. They are, however, extraordinarily challenging to identify. METHODS: To discover candidate rare variants associated with IBD, we performed whole-exome sequencing on 6 members of a pediatric-onset IBD family with multiple affected individuals. To determine whether the variants discovered in this family are also associated with nonfamilial IBD, we investigated their influence on disease in 2 large case-control (CC) series. RESULTS: We identified 2 rare variants, rs142430606 and rs200958270, both in the established IBD-susceptibility gene IL17REL, carried by all 4 affected family members and their obligate carrier parents. We then demonstrated that both variants are associated with sporadic ulcerative colitis (UC) in 2 independent data sets. For UC in CC 1: rs142430606 (odds ratio [OR] = 2.99, Padj = 0.028; minor allele frequency [MAF]cases = 0.0063, MAFcontrols = 0.0021); rs200958270 (OR = 2.61, Padj = 0.082; MAFcases = 0.0045, MAFcontrols = 0.0017). For UC in CC 2: rs142430606 (OR = 1.94, P = 0.0056; MAFcases = 0.0071, MAFcontrols = 0.0045); rs200958270 (OR = 2.08, P = 0.0028; MAFcases = 0.0071, MAFcontrols = 0.0042). CONCLUSIONS: We discover in a family and replicate in 2 CC data sets 2 rare susceptibility variants for IBD, both in IL17REL. Our results illustrate that whole-exome sequencing performed on disease-enriched families to guide association testing can be an efficient strategy for the discovery of rare disease-associated variants. We speculate that rare variants identified in families and confirmed in the general population may be important modifiers of disease risk for patients with a family history, and that genetic testing of these variants may be warranted in this patient subset.


Subject(s)
Exome/genetics , Genetic Predisposition to Disease , Inflammatory Bowel Diseases/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Interleukin-17/genetics , Adolescent , Adult , Aged , Case-Control Studies , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Pedigree , Phenotype , Prognosis , Young Adult
16.
Cancer Epidemiol Biomarkers Prev ; 24(8): 1222-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26014803

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.


Subject(s)
DNA-Binding Proteins/genetics , Genomics/methods , Nasopharyngeal Neoplasms/genetics , Carcinoma , Germ-Line Mutation , Humans , Mutation , Nasopharyngeal Carcinoma
17.
BMC Genomics ; 16: 138, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25766683

ABSTRACT

BACKGROUND: Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) associated with diseases of the colon including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). However, the functional role of many of these SNPs is largely unknown and tissue-specific resources are lacking. Expression quantitative trait loci (eQTL) mapping identifies target genes of disease-associated SNPs. This study provides a comprehensive eQTL map of distal colonic samples obtained from 40 healthy African Americans and demonstrates their relevance for GWAS of colonic diseases. RESULTS: 8.4 million imputed SNPs were tested for their associations with 16,252 expression probes representing 12,363 unique genes. 1,941 significant cis-eQTL, corresponding to 122 independent signals, were identified at a false discovery rate (FDR) of 0.01. Overall, among colon cis-eQTL, there was significant enrichment for GWAS variants for IBD (Crohn's disease [CD] and ulcerative colitis [UC]) and CRC as well as type 2 diabetes and body mass index. ERAP2, ADCY3, INPP5E, UBA7, SFMBT1, NXPE1 and REXO2 were identified as target genes for IBD-associated variants. The CRC-associated eQTL rs3802842 was associated with the expression of C11orf93 (COLCA2). Enrichment of colon eQTL near transcription start sites and for active histone marks was demonstrated, and eQTL with high population differentiation were identified. CONCLUSIONS: Through the comprehensive study of eQTL in the human colon, this study identified novel target genes for IBD- and CRC-associated genetic variants. Moreover, bioinformatic characterization of colon eQTL provides a tissue-specific tool to improve understanding of biological differences in diseases between different ethnic groups.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation/genetics , Inflammatory Bowel Diseases/genetics , Quantitative Trait Loci/genetics , Black or African American/genetics , Aged , Colorectal Neoplasms/pathology , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Inflammatory Bowel Diseases/pathology , Male , Middle Aged , Organ Specificity , Polymorphism, Single Nucleotide , Risk Factors
18.
Per Med ; 12(3): 221-229, 2015 Jun.
Article in English | MEDLINE | ID: mdl-29771652

ABSTRACT

We have recently established a biobanking and sequencing pipeline at the University of Chicago dubbed Genomics for Risk Evaluation and Anticancer Therapy in Kids. We plan to intersect family and personal history of cancer and other diseases with multidimensional genomic profiling in order to: understand how genetics may have contributed to the development of cancer for each child, and investigate the spectrum of genomic alterations within a tumor spatially (e.g., primary site vs distant metastasis) and over time (e.g., diagnosis vs relapse). This review highlights some of the practical considerations involved in creating such a program including the capacity to use our platform for multi-institutional collaborations.

19.
Int J Radiat Biol ; 90(10): 903-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24882388

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.


Subject(s)
Aging/physiology , Apoptosis/physiology , Apoptosis/radiation effects , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/radiation effects , Models, Biological , Adult , Cells, Cultured , Computer Simulation , Female , Humans , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Radiation Dosage , Reference Values , Sex Factors , Young Adult
20.
Carcinogenesis ; 35(9): 2025-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24753543

ABSTRACT

Genome-wide association studies (GWAS) in colorectal cancer (CRC) identified five regions near transforming growth factor ß-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.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Colorectal Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Black or African American , Aged , Alleles , Antigens, CD , Cadherins/genetics , Case-Control Studies , Colorectal Neoplasms/ethnology , Female , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Intercellular Signaling Peptides and Proteins/genetics , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide , Smad7 Protein/genetics , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...