9 research outputs found

    Adaptive molecular evolution of the Major Histocompatibility Complex genes, DRA and DQA, in the genus Equus

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Major Histocompatibility Complex (MHC) genes are central to vertebrate immune response and are believed to be under balancing selection by pathogens. This hypothesis has been supported by observations of extremely high polymorphism, elevated nonsynonymous to synonymous base pair substitution rates and trans-species polymorphisms at these loci. In equids, the organization and variability of this gene family has been described, however the full extent of diversity and selection is unknown. As selection is not expected to act uniformly on a functional gene, maximum likelihood codon-based models of selection that allow heterogeneity in selection across codon positions can be valuable for examining MHC gene evolution and the molecular basis for species adaptations.</p> <p>Results</p> <p>We investigated the evolution of two class II MHC genes of the Equine Lymphocyte Antigen (ELA), <it>DRA </it>and <it>DQA</it>, in the genus <it>Equus </it>with the addition of novel alleles identified in plains zebra (<it>E. quagga</it>, formerly <it>E. burchelli</it>). We found that both genes exhibited a high degree of polymorphism and inter-specific sharing of allele lineages. To our knowledge, <it>DRA </it>allelic diversity was discovered to be higher than has ever been observed in vertebrates. Evidence was also found to support a duplication of the <it>DQA </it>locus. Selection analyses, evaluated in terms of relative rates of nonsynonymous to synonymous mutations (<it>d</it><sub>N</sub><it>/d</it><sub>S</sub>) averaged over the gene region, indicated that the majority of codon sites were conserved and under purifying selection (<it>d</it><sub>N </sub><<it>d</it><sub>S</sub>). However, the most likely evolutionary codon models allowed for variable rates of selection across codon sites at both loci and, at the <it>DQA</it>, supported the hypothesis of positive selection acting on specific sites.</p> <p>Conclusions</p> <p>Observations of elevated genetic diversity and trans-species polymorphisms supported the conclusion that balancing selection may be acting on these loci. Furthermore, at the <it>DQA</it>, positive selection was occurring at antigen binding sites, suggesting that a few selected residues may play a significant role in equid immune function. Future studies in natural equid populations will be valuable for understanding the functional significance of the uniquely diverse <it>DRA </it>locus and for elucidating the mechanism maintaining diversity at these MHC loci.</p

    Association of HLA-DQ Heterodimer Residues -18Ī² and Ī²57 With Progression From Islet Autoimmunity to Diabetes in the Diabetes Prevention Trial-Type 1

    No full text
    OBJECTIVE: The purpose was to test the hypothesis that the HLA-DQĪ±Ī² heterodimer structure is related to the progression of islet autoimmunity from asymptomatic to symptomatic type 1 diabetes (T1D).RESEARCH DESIGN AND METHODS: Next-generation targeted sequencing was used to genotype HLA-DQA1-B1 class II genes in 670 subjects in the Diabetes Prevention Trial-Type 1 (DPT-1). Coding sequences were translated into DQ Ī±- and Ī²-chain amino acid residues and used in hierarchically organized haplotype (HOH) association analysis to identify motifs associated with diabetes onset.RESULTS: The opposite diabetes risks were confirmed for HLA DQA1*03:01-B1*03:02 (hazard ratio [HR] 1.36; P = 2.01 āˆ— 10-3) and DQA1*03:03-B1*03:01 (HR 0.62; P = 0.037). The HOH analysis uncovered residue -18Ī² in the signal peptide and Ī²57 in the Ī²-chain to form six motifs. DQ*VA was associated with faster (HR 1.49; P = 6.36 āˆ— 10-4) and DQ*AD with slower (HR 0.64; P = 0.020) progression to diabetes onset. VA/VA, representing DQA1*03:01-B1*03:02 (DQ8/8), had a greater HR of 1.98 (P = 2.80 āˆ— 10-3). The DQ*VA motif was associated with both islet cell antibodies (P = 0.023) and insulin autoantibodies (IAAs) (P = 3.34 āˆ— 10-3), while the DQ*AD motif was associated with a decreased IAA frequency (P = 0.015). Subjects with DQ*VA and DQ*AD experienced, respectively, increasing and decreasing trends of HbA1c levels throughout the follow-up.CONCLUSIONS: HLA-DQ structural motifs appear to modulate progression from islet autoimmunity to diabetes among at-risk relatives with islet autoantibodies. Residue -18Ī² within the signal peptide may be related to levels of protein synthesis and Ī²57 to stability of the peptide-DQab trimolecular complex

    Nine residues in HLA-DQ molecules determine with susceptibility and resistance to type 1 diabetes among young children in Sweden

    No full text
    HLA-DQ molecules account over 50% genetic risk of type 1 diabetes (T1D), but little is known about associated residues. Through next generation targeted sequencing technology and deep learning of DQ residue sequences, the aim was to uncover critical residues and their motifs associated with T1D. Our analysis uncovered (alpha a1, alpha 44, alpha 157, alpha 196) and (beta 9, beta 30, beta 57, beta 70, beta 135) on the HLA-DQ molecule. Their motifs captured all known susceptibility and resistant T1D associations. Three motifs, "DCAA-YSARD" (OR=2.10, p=1.96*10(-20)), "DQAA-YYARD" (OR=3.34, 2.69*10(-72)) and "DQDA-YYARD" (OR=3.71, 1.53*10(-6)) corresponding to DQ2.5 and DQ8.1 (the latter two motifs) associated with susceptibility. Ten motifs were significantly associated with resistance to T1D. Collectively, homozygous DQ risk motifs accounted for 43% of DQ-T1D risk, while homozygous DQ resistant motifs accounted for 25% protection to DQ-T1D risk. Of the identified nine residues five were within or near anchoring pockets of the antigenic peptide (alpha 44, beta 9, beta 30, beta 57 and beta 70), one was the N-terminal of the alpha chain (alpha a1), one in the CD4-binding region (beta 135), one in the putative cognate TCR-induced alpha beta homodimerization process (alpha 157), and one in the intra-membrane domain of the alpha chain (alpha 196). Finding these critical residues should allow investigations of fundamental properties of host immunity that underlie tolerance to self and organ-specific autoimmunity.Funding Agencies|Lund University; National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)United States Department of Health &amp; Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Diabetes &amp; Digestive &amp; Kidney Diseases (NIDDK) [1R01DK117276]; European Foundation for the Study of Diabetes (EFSD) Clinical Research Grants Programme 2013; Swedish Child Diabetes Foundation (Barndiabetesfonden); Swedish Foundation for Strategic ResearchSwedish Foundation for Strategic Research [IRC15-0067]; Swedish Research Council, Strategic Research AreaSwedish Research Council [2009-1039]; Skane County Council for Research and Development; Swedish Association of Local Authorities and Regions (SKL)</p

    Motifs of Three HLA-DQ Amino Acid Residues (Ī±44, Ī²57, Ī²135) Capture Full Association With the Risk of Type 1 Diabetes in DQ2 and DQ8 Children

    No full text
    HLA-DQA1 and -DQB1 are strongly associated with type 1 diabetes (T1D), and DQ8.1 and DQ2.5 are major risk haplotypes. Next-generation targeted sequencing of HLA-DQA1 and -DQB1 in Swedish newly diagnosed 1- to 18 year-old patients (n = 962) and control subjects (n = 636) was used to construct abbreviated DQ haplotypes, converted into amino acid (AA) residues, and assessed for their associations with T1D. A hierarchically organized haplotype (HOH) association analysis allowed 45 unique DQ haplotypes to be categorized into seven clusters. The DQ8/9 cluster included two DQ8.1 risk and the DQ9 resistant haplotypes, and the DQ2 cluster included the DQ2.5 risk and DQ2.2 resistant haplotypes. Within each cluster, HOH found residues Ī±44Q (odds ratio [OR] 3.29, P = 2.38 * 10-85) and Ī²57A (OR 3.44, P = 3.80 * 10-84) to be associated with T1D in the DQ8/9 cluster representing all ten residues (Ī±22, Ī±23, Ī±44, Ī±49, Ī±51, Ī±53, Ī±54, Ī±73, Ī±184, Ī²57) due to complete linkage disequilibrium (LD) of Ī±44 with eight such residues. Within the DQ2 cluster and due to LD, HOH analysis found Ī±44C and Ī²135D to share the risk for T1D (OR 2.10, P = 1.96 * 10-20). The motif "QAD" of Ī±44, Ī²57, and Ī²135 captured the T1D risk association of DQ8.1 (OR 3.44, P = 3.80 * 10-84), and the corresponding motif "CAD" captured the risk association of DQ2.5 (OR 2.10, P = 1.96 * 10-20). Two risk associations were related to GAD65 autoantibody (GADA) and IA-2 autoantibody (IA-2A) but in opposite directions. CAD was positively associated with GADA (OR 1.56, P = 6.35 * 10-8) but negatively with IA-2A (OR 0.59, P = 6.55 * 10-11). QAD was negatively associated with GADA (OR 0.88; P = 3.70 * 10-3) but positively with IA-2A (OR 1.64; P = 2.40 * 10-14), despite a single difference at Ī±44. The residues are found in and around anchor pockets 1 and 9, as potential T-cell receptor contacts, in the areas for CD4 binding and putative homodimer formation. The identification of three HLA-DQ AAs (Ī±44, Ī²57, Ī²135) conferring T1D risk should sharpen functional and translational studies

    Next Generation HLA Sequence Analysis Uncovers Seven HLA-DQ Amino Acid Residues and Six Motifs Resistant to Childhood Type 1 Diabetes

    No full text
    HLA-DQA1 and -DQB1 genes have significant and potentially causal associations with autoimmune type 1 diabetes (T1D). To follow on the earlier analysis on high-risk HLA-DQ2.5 and DQ8.1, the current analysis uncovers seven residues (Ī±a1, Ī±157, Ī±196, Ī²9, Ī²30, Ī²57, Ī²70) that are resistant to T1D among subjects with DQ4, 5, 6 and 7 resistant DQ haplotypes. These seven residues form 13 common motifs; six motifs are significantly resistant, six motifs have modest or no associations (p-values>0.05), and one motif has 7 copies observed among controls only. The motif "DAAFYDG", "DAAYHDG" and "DAAYYDR" have significant resistance to T1D (OR = 0.03, 0.25 and 0.18, p-value = 6.11*10-24, 3.54*10-15 and 1.03*10-21, respectively). Remarkably, a change of a single residue from the motif "DAAYH D G" to "DAAYH S G" (D to S at Ī²57) alters the resistance potential, from resistant motif (OR = 0.15, p-value = 3.54*10-15) to a neutral motif (p-value = 0.183), the change of which was significant (Fisher's p-value = 0.0065). The extended set of linked residues associated with T1D resistance and unique to each cluster of HLA-DQ haplotypes represents facets of all known features and functions of these molecules: antigenic peptide binding, pMHCII complex stability, Ī²167-169 RGD loop, TCR binding, formation of homodimer of alpha-beta heterodimers, and cholesterol binding in the cell membrane rafts. Identifications of these residues is a novel understanding of resistant DQ associations with T1D. Our analyses endow potential molecular approaches to identify immunological mechanisms that control disease susceptibility or resistance to provide novel targets for immunotherapeutic strategies

    Eleven Amino Acids of HLA-DRB1 and Fifteen Amino Acids of HLA-DRB3, 4 and 5 Include Potentially "Causal Residues" Responsible for the Risk of Childhood Type 1 Diabetes

    No full text
    Next generation targeted sequencing of HLA-DRB1, -DRB3, -DRB4 and -DRB5 (abbreviated as DRB345) provides high resolution of functional variant positions to investigate their associations with type 1 diabetes risk and with autoantibodies against insulin (IAA), GAD65 (GADA), IA-2 (IA-2A) or ZnT8 (ZnT8A). To overcome exceptional DR sequence complexity due to high polymorphisms and extended linkage-disequilibrium among the DR loci, we apply a novel recursive organizer (ROR) to discover disease-associated amino acid residues. ROR distills disease associated DR sequences down and identifies eleven residues of DRB1, sequences of which retain all significant associations observed by DR genes. Further, all eleven residues locate under/adjoining the peptide binding groove of DRB1, suggesting a plausible functional mechanism through peptide binding. In addition, 15 residues of DRB345, located respectively in the Ī²50-55 homodimerization patch and in face of the molecule shown to interact with and bind to the accessory molecule CD4, retain their significant disease associations. Further ROR analysis of DR associations with autoantibodies finds DRB1 residues significantly associated with ZnT8A and DRB345-residues with GADA. The strongest association is between four residues (Ļ‡14, Ī²25, Ī²71 and Ī²73) and IA-2A, in which a sequence "ERKA" confers a risk association (OR=2.15, p-value=10-18), and another sequence "ERKG" confers a protective association (OR=0.59, p-value=10-11), despite a difference of only one amino acid. As motifs of identified residues capture potentially causal DR associations with type 1 diabetes, this list of residuals is expected to include corresponding causal residues in this study population

    The KAG motif of HLA-DRB1 (Ī²71, Ī²74, Ī²86) predicts seroconversion and development of type 1 diabetes

    No full text
    Background: HLA-DR4, a common antigen of HLA-DRB1, has multiple subtypes that are strongly associated with risk of type 1 diabetes (T1D); however, some are risk neutral or resistant. The pathobiological mechanism of HLA-DR4 subtypes remains to be elucidated. Methods: We used a population-based case-control study of T1D (962 patients and 636 controls) to decipher genetic associations of HLA-DR4 subtypes and specific residues with susceptibility to T1D. Using a birth cohort of 7865 children with periodically measured islet autoantibodies (GADA, IAA or IA-2A), we proposed to validate discovered genetic associations with a totally different study design and time-to-seroconversions prior to clinical onset of T1D. A novel analytic strategy hierarchically organized the HLA-DRB1 alleles by sequence similarity and identified critical amino acid residues by minimizing local genomic architecture and higher-order interactions. Findings: Three amino acid residues of HLA-DRB1 (Ī²71, Ī²74, Ī²86) were found to be predictive of T1D risk in the population-based study. The ā€œKAGā€ motif, corresponding to HLA-DRB1Ɨ04:01, was most strongly associated with T1D risk ([O]dds [R]atio=3.64, pĀ =Ā 3.19Ā Ć—Ā 10āˆ’64). Three less frequent motifs (ā€œEAVā€, ORĀ =Ā 2.55, pĀ =Ā 0.025; ā€œRAGā€, ORĀ =Ā 1.93, pĀ =Ā 0.043; and ā€œRAVā€, ORĀ =Ā 1.56, pĀ =Ā 0.003) were associated with T1D risk, while two motifs (ā€œREGā€ and ā€œREVā€) were equally protective (ORĀ =Ā 0.11, pĀ =Ā 4.23Ā Ć—Ā 10āˆ’4). In an independent birth cohort of HLA-DR3 and HLA-DR4 subjects, those having the ā€œKAGā€ motif had increased risk for time-to-seroconversion (Hazard RatioĀ =Ā 1.74, pĀ =Ā 6.51Ā Ć—Ā 10āˆ’14) after adjusting potential confounders. Interpretations: DNA sequence variation in HLA-DRB1 at positions Ī²71, Ī²74, and Ī²86 are non-conservative (Ī²74 Aā†’E, Ī²71 E vs K vs R and Ī²86Ā G vs V). They result in substantial differences in peptide antigen anchor pocket preferences at p1, p4 and potentially neighboring regions such as pocket p7. Differential peptide antigen binding is likely to be affected. These sequence substitutions may account for most of the HLA-DR4 contribution to T1D risk as illustrated in two HLA-peptide model complexes of the T1D autoantigens preproinsulin and GAD65. Funding: National Institute of Diabetes and Digestive and Kidney Diseases and the Swedish Child Diabetes Foundation and the Swedish Research Council
    corecore