Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Allergy Clin Immunol ; 139(2): 634-642.e5, 2017 02.
Article in English | MEDLINE | ID: mdl-27522155

ABSTRACT

BACKGROUND: Signaling through the T-cell receptor (TCR) is critical for T-cell development and function. Linker for activation of T cells (LAT) is a transmembrane adaptor signaling molecule that is part of the TCR complex and essential for T-cell development, as demonstrated by LAT-deficient mice, which show a complete lack of peripheral T cells. OBJECTIVE: We describe a pedigree affected by a severe combined immunodeficiency phenotype with absent T cells and normal B-cell and natural killer cell numbers. A novel homozygous frameshift mutation in the gene encoding for LAT was identified in this kindred. METHODS: Genetic, molecular, and functional analyses were used to identify and characterize the LAT defect. Clinical and immunologic analysis of patients was also performed and reported. RESULTS: Homozygosity mapping was used to identify potential defective genes. Sanger sequencing of the LAT gene showed a mutation that resulted in a premature stop codon and protein truncation leading to complete loss of function and loss of expression of LAT in the affected family members. We also demonstrate loss of LAT expression and lack of TCR signaling restoration in LAT-deficient cell lines reconstituted with a synthetic LAT gene bearing this severe combined immunodeficiency mutation. CONCLUSION: For the first time, the results of this study show that inherited LAT deficiency should be considered in patients with combined immunodeficiency with T-cell abnormalities.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Sequence Deletion/genetics , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Calcium Signaling/genetics , Cell Differentiation , Consanguinity , Female , Genotype , Homozygote , Humans , Jurkat Cells , Lymphocyte Activation , Male , Membrane Proteins/genetics , Pakistan , Pedigree , Receptors, Antigen, T-Cell/genetics , Transgenes/genetics
2.
N Engl J Med ; 364(7): 616-26, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21323541

ABSTRACT

BACKGROUND: Idiopathic membranous nephropathy is a major cause of the nephrotic syndrome in adults, but its etiologic basis is not fully understood. We investigated the genetic basis of biopsy-proven cases of idiopathic membranous nephropathy in a white population. METHODS: We performed independent genomewide association studies of single-nucleotide polymorphisms (SNPs) in patients with idiopathic membranous nephropathy from three populations of white ancestry (75 French, 146 Dutch, and 335 British patients). The patients were compared with racially matched control subjects; population stratification and quality controls were carried out according to standard criteria. Associations were calculated by means of a chi-square basic allele test; the threshold for significance was adjusted for multiple comparisons (with the Bonferroni method). RESULTS: In a joint analysis of data from the 556 patients studied (398 men), we identified significant alleles at two genomic loci associated with idiopathic membranous nephropathy. Chromosome 2q24 contains the gene encoding M-type phospholipase A(2) receptor (PLA(2)R1) (SNP rs4664308, P=8.6×10(-29)), previously shown to be the target of an autoimmune response. Chromosome 6p21 contains the gene encoding HLA complex class II HLA-DQ alpha chain 1 (HLA-DQA1) (SNP rs2187668, P=8.0×10(-93)). The association with HLA-DQA1 was significant in all three populations (P=1.8×10(-9), P=5.6×10(-27), and P=5.2×10(-36) in the French, Dutch, and British groups, respectively). The odds ratio for idiopathic membranous nephropathy with homozygosity for both risk alleles was 78.5 (95% confidence interval, 34.6 to 178.2). CONCLUSIONS: An HLA-DQA1 allele on chromosome 6p21 is most closely associated with idiopathic membranous nephropathy in persons of white ancestry. This allele may facilitate an autoimmune response against targets such as variants of PLA2R1. Our findings suggest a basis for understanding this disease and illuminate how adaptive immunity is regulated by HLA.


Subject(s)
Genome-Wide Association Study , Glomerulonephritis, Membranous/genetics , HLA-DQ Antigens/genetics , Polymorphism, Single Nucleotide , Receptors, Phospholipase A2/genetics , Alleles , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 6 , Europe , Female , Genotype , HLA-DQ alpha-Chains , Humans , Male , Odds Ratio , White People/genetics
3.
Nat Genet ; 37(10): 1119-24, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142236

ABSTRACT

Intrauterine growth retardation is caused by maternal, fetal or placental factors that result in impaired endovascular trophoblast invasion and reduced placental perfusion. Although various causes of intrauterine growth retardation have been identified, most cases remain unexplained. Studying 29 families with 3-M syndrome (OMIM 273750), an autosomal recessive condition characterized by severe pre- and postnatal growth retardation, we first mapped the underlying gene to chromosome 6p21.1 and then identified 25 distinct mutations in the gene cullin 7 (CUL7). CUL7 assembles an E3 ubiquitin ligase complex containing Skp1, Fbx29 (also called Fbw8) and ROC1 and promotes ubiquitination. Using deletion analysis, we found that CUL7 uses its central region to interact with the Skp1-Fbx29 heterodimer. Functional studies indicated that the 3-M-associated CUL7 nonsense and missense mutations R1445X and H1464P, respectively, render CUL7 deficient in recruiting ROC1. These results suggest that impaired ubiquitination may have a role in the pathogenesis of intrauterine growth retardation in humans.


Subject(s)
Chromosomes, Human, Pair 6/genetics , Cullin Proteins/genetics , Fetal Growth Retardation/genetics , Carrier Proteins/metabolism , Child , Chromosome Mapping , Codon, Nonsense , DNA Mutational Analysis , Female , Homozygote , Humans , Male , Mutation, Missense , Protein Interaction Mapping , Protein Structure, Tertiary , S-Phase Kinase-Associated Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Sequence Deletion , Syndrome
4.
Dev Biol ; 340(2): 369-80, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20122914

ABSTRACT

22q11 deletion syndrome (22q11DS) is characterised by aberrant development of the pharyngeal apparatus and the heart with haploinsufficiency of the transcription factor TBX1 being considered the major underlying cause of the disease. Tbx1 mutations in mouse phenocopy the disorder. In order to identify the transcriptional dysregulation in Tbx1-expressing lineages we optimised fluorescent-activated cell sorting of beta-galactosidase expressing cells (FACS-Gal) to compare the expression profile of Df1/Tbx1(lacZ) (effectively Tbx1 null) and Tbx1 heterozygous cells isolated from mouse embryos. Hes1, a major effector of Notch signalling, was identified as downregulated in Tbx1(-)(/)(-) mutants. Hes1 mutant mice exhibited a partially penetrant range of 22q11DS-like defects including pharyngeal arch artery (PAA), outflow tract, craniofacial and thymic abnormalities. Similar to Tbx1 mice, conditional mutagenesis revealed that Hes1 expression in embryonic pharyngeal ectoderm contributes to thymus and pharyngeal arch artery development. These results suggest that Hes1 acts downstream of Tbx1 in the morphogenesis of pharyngeal-derived structures.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Branchial Region/embryology , Heart/embryology , Homeodomain Proteins/metabolism , Sequence Deletion , Thymus Gland/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Branchial Region/metabolism , Chromosomes/genetics , Embryo, Mammalian/metabolism , Homeodomain Proteins/genetics , In Situ Hybridization , Mice , Mice, Knockout , Syndrome , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Thymus Gland/metabolism , Transcription Factor HES-1 , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
Stroke ; 42(10): 2726-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21940970

ABSTRACT

BACKGROUND AND PURPOSE: Ischemic stroke has a strong familial component to risk. The Siblings With Ischemic Stroke Study (SWISS) is a genome-wide, family-based analysis that included use of imputed genotypes. The Siblings With Ischemic Stroke Study was conducted to examine the associations between single-nucleotide polymorphisms (SNPs) and risk of stroke and stroke subtypes within pairs. METHODS: The Siblings With Ischemic Stroke Study enrolled 312 probands with ischemic stroke from 70 US and Canadian centers. Affected siblings were ascertained by centers and confirmed by central record review; unaffected siblings were ascertained by telephone contact. Ischemic stroke was subtyped according to Trial of Org 10172 in Acute Stroke Treatment criteria. Genotyping was performed with an Illumina 610 quad array (probands) and an Illumina linkage V array (affected siblings). SNPs were imputed by using 1000 Genomes Project data and MACH software. Family-based association analyses were conducted by using the sibling transmission-disequilibrium test. RESULTS: For all pairs, the correlation of age at stroke within pairs of affected siblings was r=0.83 (95% CI, 0.78-0.86; P<2.2×10(-16)). The correlation did not differ substantially by subtype. The concordance of stroke subtypes among affected pairs was 33.8% (kappa=0.13; P=5.06×10(-4)) and did not differ by age at stroke in the proband. Although no SNP achieved genome-wide significance for risk of ischemic stroke, there was clustering of the most associated SNPs on chromosomes 3p (neuronal nitric oxide synthase) and 6p. CONCLUSIONS: Stroke subtype and age at stroke in affected sibling pairs exhibit significant clustering. No individual SNP reached genome-wide significance. However, 2 promising candidate loci were identified, including 1 that contains neuronal nitric oxide synthase, although these risk loci warrant further examination in larger sample collections.


Subject(s)
Brain Ischemia/genetics , Genetic Predisposition to Disease , Siblings , Stroke/genetics , Adult , Aged , Aged, 80 and over , Female , Genetic Loci , Genetic Testing , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
6.
Methods ; 52(3): 248-54, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20434562

ABSTRACT

Aberrant DNA methylation of promoter and other genomic regions can lead to changes in gene expression, such as over-expression of oncogenes and the silencing of tumour suppressor genes in the context of cancer. Ability to accurately assess the DNA methylation status is therefore of great importance in health and disease. Recently, various platforms for genome-wide analysis of promoter DNA methylation have been developed, including the Illumina Infinium platform. While some of these platforms can be used with formalin-fixed paraffin-embedded (FFPE) tissue, no protocol has yet been developed for the analysis of FFPE-derived DNA on the Infinium platform using the HumanMethylation27 BeadChip which interrogates 27,578 cytosine-guanine (CpG) dinucleotide sites, selected predominantly from the promoter regions of 14,000 annotated genes. As FFPE preservation has been the method of choice for the archiving of clinical samples, the ability to analyse such samples opens the possibility to study large numbers of clinically well annotated samples which can be expected to lead to more powerful and robust data, particularly for rare cancers. Here, we describe a protocol for the analysis of FFPE samples using the Infinium HumanMethylation27 BeadChip.


Subject(s)
DNA Methylation , Genome, Human/genetics , Oligonucleotide Array Sequence Analysis/methods , Paraffin Embedding , Tissue Fixation , Formaldehyde , Humans
7.
Hum Mol Genet ; 17(14): 2150-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18407919

ABSTRACT

Homozygous loss-of-function mutations in the transcription factor LHX3 have been associated with hypopituitarism with structural anterior pituitary defects and cervical abnormalities with or without restricted neck rotation. We report two novel recessive mutations in LHX3 in four patients from two unrelated pedigrees. Clinical evaluation revealed that all four patients exhibit varying degrees of bilateral sensorineural hearing loss, which has not been previously reported in association with LHX3 mutations, in addition to hypopituitarism including adrenocorticotropic hormone deficiency and an unusual skin and skeletal phenotype in one family. Furthermore, re-evaluation of three patients previously described with LHX3 mutations showed they also exhibit varying degrees of bilateral sensorineural hearing loss. We have investigated a possible role for LHX3 in inner ear development in humans using in situ hybridization of human embryonic and fetal tissue. LHX3 is expressed in defined regions of the sensory epithelium of the developing inner ear in a pattern overlapping that of SOX2, which precedes the onset of LHX3 expression and is known to be required for inner ear and pituitary development in both mice and humans. Moreover, we show that SOX2 is capable of binding to and activating transcription of the LHX3 proximal promoter in vitro. This study therefore extends the phenotypic spectrum associated with LHX3 mutations to encompass variable sensorineural hearing loss and suggests a possible interaction between LHX3 and SOX2 likely to be important for development of both the inner ear and the anterior pituitary in human embryonic development.


Subject(s)
Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Hypopituitarism/genetics , Mutation , Adolescent , Animals , Base Sequence , CHO Cells , Child , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ear, Inner/embryology , Ear, Inner/growth & development , Ear, Inner/metabolism , Embryo, Mammalian/metabolism , Female , Gene Expression , HMGB Proteins/genetics , HMGB Proteins/metabolism , Hearing Loss, Sensorineural/embryology , Hearing Loss, Sensorineural/metabolism , Homeodomain Proteins/metabolism , Humans , Hypopituitarism/embryology , Hypopituitarism/metabolism , Infant , LIM-Homeodomain Proteins , Male , Mice , Polymorphism, Single Nucleotide , SOXB1 Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
8.
Leukemia ; 33(12): 2817-2829, 2019 12.
Article in English | MEDLINE | ID: mdl-31138842

ABSTRACT

Subtype-specific leukemia oncogenes drive aberrant gene expression profiles that converge on common essential mediators to ensure leukemia self-renewal and inhibition of differentiation. The transcription factor c-MYB functions as one such mediator in a diverse range of leukemias. Here we show for the first time that transcriptional repression of myeloid differentiation associated c-MYB target genes in AML is enforced by the AAA+ ATPase RUVBL2. Silencing RUVBL2 expression resulted in increased binding of c-MYB to these loci and their transcriptional activation. RUVBL2 inhibition resulted in AML cell apoptosis and severely impaired disease progression of established AML in engrafted mice. In contrast, such inhibition had little impact on normal hematopoietic progenitor differentiation. These data demonstrate that RUVBL2 is essential for the oncogenic function of c-MYB in AML by governing inhibition of myeloid differentiation. They also indicate that targeting the control of c-MYB function by RUVBL2 is a promising approach to developing future anti-AML therapies.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Helicases/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-myb/genetics , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cell Line, Tumor , DNA Helicases/genetics , Disease Models, Animal , Disease Progression , Gene Knockdown Techniques , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Protein Binding , Proto-Oncogene Proteins c-myb/metabolism , Xenograft Model Antitumor Assays
9.
Eur J Hum Genet ; 27(7): 1121-1133, 2019 07.
Article in English | MEDLINE | ID: mdl-30886340

ABSTRACT

Recent studies have demonstrated genetic differences between monozygotic (MZ) twins. To test the hypothesis that early post-twinning mutational events associate with phenotypic discordance, we investigated a cohort of 13 twin pairs (n = 26) discordant for various clinical phenotypes using whole-exome sequencing and screened for copy number variation (CNV). We identified a de novo variant in PLCB1, a gene involved in the hydrolysis of lipid phosphorus in milk from dairy cows, associated with lactase non-persistence, and a variant in the mitochondrial complex I gene MT-ND5 associated with amyotrophic lateral sclerosis (ALS). We also found somatic variants in multiple genes (TMEM225B, KBTBD3, TUBGCP4, TFIP11) in another MZ twin pair discordant for ALS. Based on the assumption that discordance between twins could be explained by a common variant with variable penetrance or expressivity, we screened the twin samples for known pathogenic variants that are shared and identified a rare deletion overlapping ARHGAP11B, in the twin pair manifesting with either schizotypal personality disorder or schizophrenia. Parent-offspring trio analysis was implemented for two twin pairs to assess potential association of variants of parental origin with susceptibility to disease. We identified a de novo variant in RASD2 shared by 8-year-old male twins with a suspected diagnosis of autism spectrum disorder (ASD) manifesting as different traits. A de novo CNV duplication was also identified in these twins overlapping CD38, a gene previously implicated in ASD. In twins discordant for Tourette's syndrome, a paternally inherited stop loss variant was detected in AADAC, a known candidate gene for the disorder.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Autism Spectrum Disorder/genetics , Base Sequence , DNA Copy Number Variations , Sequence Deletion , Tourette Syndrome/genetics , Twins, Monozygotic/genetics , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Exome Sequencing
10.
Cancer Res ; 73(18): 5834-44, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23887970

ABSTRACT

Brain tumors are thought to originate from stem/progenitor cell populations that acquire specific genetic mutations. Although current preclinical models have relevance to human pathogenesis, most do not recapitulate the histogenesis of the human disease. Recently, a large series of human gliomas and medulloblastomas were analyzed for genetic signatures of prognosis and therapeutic response. Using a mouse model system that generates three distinct types of intrinsic brain tumors, we correlated RNA and protein expression levels with human brain tumors. A combination of genetic mutations and cellular environment during tumor propagation defined the incidence and phenotype of intrinsic murine tumors. Importantly, in vitro passage of cancer stem cells uniformly promoted a glial expression profile in culture and in brain tumors. Gene expression profiling revealed that experimental gliomas corresponded to distinct subclasses of human glioblastoma, whereas experimental supratentorial primitive neuroectodermal tumors (sPNET) correspond to atypical teratoid/rhabdoid tumor (AT/RT), a rare childhood tumor.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain/metabolism , Cell Lineage , Gene Expression Profiling , Glioma/genetics , Neoplastic Stem Cells/pathology , Animals , Brain/cytology , Brain Neoplasms/classification , Brain Neoplasms/pathology , Glioma/classification , Glioma/pathology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/physiology , Phenotype , Proteins/physiology , RNA, Messenger/genetics , RNA, Untranslated , Real-Time Polymerase Chain Reaction , Retinoblastoma Protein/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/physiology
11.
BMJ Open ; 2: e000411, 2012.
Article in English | MEDLINE | ID: mdl-22223843

ABSTRACT

Background The mitochondrial DNA mutation m.1555A>G predisposes to permanent idiosyncratic aminoglycoside-induced deafness that is independent of dose. Research suggests that in some families, m.1555A>G may cause non-syndromic deafness, without aminoglycoside exposure, as well as reduced hearing thresholds with age (age-related hearing loss). Objectives To determine whether adults with m.1555A>G have impaired hearing, a factor that would inform the cost-benefit argument for genetic testing prior to aminoglycoside administration. Design Population-based cohort study. Setting UK. Participants Individuals from the British 1958 birth cohort. Measurements Hearing thresholds at 1 and 4 kHz at age 44-45 years; m.1555A>G genotyping. Results 19 of 7350 individuals successfully genotyped had the m.1555A>G mutation, giving a prevalence of 0.26% (95% CI 0.14% to 0.38%) or 1 in 385 (95% CI 1 in 714 to 1 in 263). There was no significant difference in hearing thresholds between those with and without the mutation. Single-nucleotide polymorphism analysis indicated that the mutation has arisen on a number of different mitochondrial haplogroups. Limitations No data were collected on aminoglycoside exposure. For three subjects, hearing thresholds could not be predicted because information required for modelling was missing. Conclusions In this cohort, hearing in those with m.1555A>G is not significantly different from the general population and appears to be preserved at least until 44-45 years of age. Unbiased ascertainment of mutation carriers provides no evidence that this mutation alone causes non-syndromic hearing impairment in the UK. The findings lend weight to arguments for genetic testing for this mutation prior to aminoglycoside administration, as hearing in susceptible individuals is expected to be preserved well into adult life. Since global use of aminoglycosides is likely to increase, development of a rapid test is a priority.

12.
PLoS One ; 6(9): e23161, 2011.
Article in English | MEDLINE | ID: mdl-21957438

ABSTRACT

INTRODUCTION: Familial aggregation of ischemic stroke derives from shared genetic and environmental factors. We present a meta-analysis of genome-wide association scans (GWAS) from 3 cohorts to identify the contribution of common variants to ischemic stroke risk. METHODS: This study involved 1464 ischemic stroke cases and 1932 controls. Cases were genotyped using the Illumina 610 or 660 genotyping arrays; controls, with Illumina HumanHap 550Kv1 or 550Kv3 genotyping arrays. Imputation was performed with the 1000 Genomes European ancestry haplotypes (August 2010 release) as a reference. A total of 5,156,597 single-nucleotide polymorphisms (SNPs) were incorporated into the fixed effects meta-analysis. All SNPs associated with ischemic stroke (P<1×10(-5)) were incorporated into a multivariate risk profile model. RESULTS: No SNP reached genome-wide significance for ischemic stroke (P<5×10(-8)). Secondary analysis identified a significant cumulative effect for age at onset of stroke (first versus fifth quintile of cumulative profiles based on SNPs associated with late onset, ß = 14.77 [10.85,18.68], P = 5.5×10(-12)), as well as a strong effect showing increased risk across samples with a high propensity for stroke among samples with enriched counts of suggestive risk alleles (P<5×10(-6)). Risk profile scores based only on genomic information offered little incremental prediction. DISCUSSION: There is little evidence of a common genetic variant contributing to moderate risk of ischemic stroke. Quintiles based on genetic loading of alleles associated with a younger age at onset of ischemic stroke revealed a significant difference in age at onset between those in the upper and lower quintiles. Using common variants from GWAS and imputation, genomic profiling remains inferior to family history of stroke for defining risk. Inclusion of genomic (rare variant) information may be required to improve clinical risk profiling.


Subject(s)
Brain Ischemia/complications , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genomics , Internationality , Stroke/complications , Stroke/genetics , Age of Onset , Aged , Cohort Studies , Female , Humans , Male , Polymorphism, Single Nucleotide/genetics , Stroke/epidemiology , Stroke/etiology
13.
Nat Genet ; 43(12): 1193-201, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057235

ABSTRACT

Using variants from the 1000 Genomes Project pilot European CEU dataset and data from additional resequencing studies, we densely genotyped 183 non-HLA risk loci previously associated with immune-mediated diseases in 12,041 individuals with celiac disease (cases) and 12,228 controls. We identified 13 new celiac disease risk loci reaching genome-wide significance, bringing the number of known loci (including the HLA locus) to 40. We found multiple independent association signals at over one-third of these loci, a finding that is attributable to a combination of common, low-frequency and rare genetic variants. Compared to previously available data such as those from HapMap3, our dense genotyping in a large sample collection provided a higher resolution of the pattern of linkage disequilibrium and suggested localization of many signals to finer scale regions. In particular, 29 of the 54 fine-mapped signals seemed to be localized to single genes and, in some instances, to gene regulatory elements. Altogether, we define the complex genetic architecture of the risk regions of and refine the risk signals for celiac disease, providing the next step toward uncovering the causal mechanisms of the disease.


Subject(s)
Celiac Disease/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Chromosome Mapping , Gene Frequency , Genetic Loci , Genome-Wide Association Study , Haplotypes , Humans , Linkage Disequilibrium , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL