Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Cell ; 153(3): 707-20, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23622250

ABSTRACT

The genetics of complex disease produce alterations in the molecular interactions of cellular pathways whose collective effect may become clear through the organized structure of molecular networks. To characterize molecular systems associated with late-onset Alzheimer's disease (LOAD), we constructed gene-regulatory networks in 1,647 postmortem brain tissues from LOAD patients and nondemented subjects, and we demonstrate that LOAD reconfigures specific portions of the molecular interaction structure. Through an integrative network-based approach, we rank-ordered these network structures for relevance to LOAD pathology, highlighting an immune- and microglia-specific module that is dominated by genes involved in pathogen phagocytosis, contains TYROBP as a key regulator, and is upregulated in LOAD. Mouse microglia cells overexpressing intact or truncated TYROBP revealed expression changes that significantly overlapped the human brain TYROBP network. Thus the causal network structure is a useful predictor of response to gene perturbations and presents a framework to test models of disease mechanisms underlying LOAD.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Gene Regulatory Networks , Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Animals , Bayes Theorem , Brain/pathology , Humans , Membrane Proteins/metabolism , Mice , Microglia/metabolism
2.
Am J Hum Genet ; 109(5): 885-899, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35325614

ABSTRACT

Genome-wide association studies (GWASs) of Huntington disease (HD) have identified six DNA maintenance gene loci (among others) as modifiers and implicated a two step-mechanism of pathogenesis: somatic instability of the causative HTT CAG repeat with subsequent triggering of neuronal damage. The largest studies have been limited to HD individuals with a rater-estimated age at motor onset. To capitalize on the wealth of phenotypic data in several large HD natural history studies, we have performed algorithmic prediction by using common motor and cognitive measures to predict age at other disease landmarks as additional phenotypes for GWASs. Combined with imputation with the Trans-Omics for Precision Medicine reference panel, predictions using integrated measures provided objective landmark phenotypes with greater power to detect most modifier loci. Importantly, substantial differences in the relative modifier signal across loci, highlighted by comparing common modifiers at MSH3 and FAN1, revealed that individual modifier effects can act preferentially in the motor or cognitive domains. Individual components of the DNA maintenance modifier mechanisms may therefore act differentially on the neuronal circuits underlying the corresponding clinical measures. In addition, we identified additional modifier effects at the PMS1 and PMS2 loci and implicated a potential second locus on chromosome 7. These findings indicate that broadened discovery and characterization of HD genetic modifiers based on additional quantitative or qualitative phenotypes offers not only the promise of in-human validated therapeutic targets but also a route to dissecting the mechanisms and cell types involved in both the somatic instability and toxicity components of HD pathogenesis.


Subject(s)
Huntington Disease , Cognition , DNA , Genome-Wide Association Study , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Trinucleotide Repeat Expansion
3.
Hum Mol Genet ; 30(3-4): 135-148, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33432339

ABSTRACT

Huntington's disease pathogenesis involves a genetic gain-of-function toxicity mechanism triggered by the expanded HTT CAG repeat. Current therapeutic efforts aim to suppress expression of total or mutant huntingtin, though the relationship of huntingtin's normal activities to the gain-of-function mechanism and what the effects of huntingtin-lowering might be are unclear. Here, we have re-investigated a rare family segregating two presumed HTT loss-of-function (LoF) variants associated with the developmental disorder, Lopes-Maciel-Rodan syndrome (LOMARS), using whole-genome sequencing of DNA from cell lines, in conjunction with analysis of mRNA and protein expression. Our findings correct the muddled annotation of these HTT variants, reaffirm they are the genetic cause of the LOMARS phenotype and demonstrate that each variant is a huntingtin hypomorphic mutation. The NM_002111.8: c.4469+1G>A splice donor variant results in aberrant (exon 34) splicing and severely reduced mRNA, whereas, surprisingly, the NM_002111.8: c.8157T>A NP_002102.4: Phe2719Leu missense variant results in abnormally rapid turnover of the Leu2719 huntingtin protein. Thus, although rare and subject to an as yet unknown LoF intolerance at the population level, bona fide HTT LoF variants can be transmitted by normal individuals leading to severe consequences in compound heterozygotes due to huntingtin deficiency.


Subject(s)
Gene Expression Regulation , Huntingtin Protein/genetics , Mutation , Neurodevelopmental Disorders/genetics , Amino Acid Sequence , Cell Line , Child , Child, Preschool , Female , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Loss of Function Mutation , Male , Mutation, Missense , Neurodevelopmental Disorders/metabolism , Pedigree , Phenotype , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA
4.
Am J Hum Genet ; 107(1): 96-110, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32589923

ABSTRACT

A recent genome-wide association study of Huntington disease (HD) implicated genes involved in DNA maintenance processes as modifiers of onset, including multiple genome-wide significant signals in a chr15 region containing the DNA repair gene Fanconi-Associated Nuclease 1 (FAN1). Here, we have carried out detailed genetic, molecular, and cellular investigation of the modifiers at this locus. We find that missense changes within or near the DNA-binding domain (p.Arg507His and p.Arg377Trp) reduce FAN1's DNA-binding activity and its capacity to rescue mitomycin C-induced cytotoxicity, accounting for two infrequent onset-hastening modifier signals. We also idenified a third onset-hastening modifier signal whose mechanism of action remains uncertain but does not involve an amino acid change in FAN1. We present additional evidence that a frequent onset-delaying modifier signal does not alter FAN1 coding sequence but is associated with increased FAN1 mRNA expression in the cerebral cortex. Consistent with these findings and other cellular overexpression and/or suppression studies, knockout of FAN1 increased CAG repeat expansion in HD-induced pluripotent stem cells. Together, these studies support the process of somatic CAG repeat expansion as a therapeutic target in HD, and they clearly indicate that multiple genetic variations act by different means through FAN1 to influence HD onset in a manner that is largely additive, except in the rare circumstance that two onset-hastening alleles are present. Thus, an individual's particular combination of FAN1 haplotypes may influence their suitability for HD clinical trials, particularly if the therapeutic agent aims to reduce CAG repeat instability.


Subject(s)
Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Huntington Disease/genetics , Multifunctional Enzymes/genetics , Cell Line , Genome-Wide Association Study/methods , HEK293 Cells , Haplotypes/genetics , Humans , Polymorphism, Single Nucleotide/genetics
5.
Hum Mol Genet ; 29(18): 3044-3053, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32876667

ABSTRACT

Recent genome-wide association studies of age-at-onset in Huntington's disease (HD) point to distinct modes of potential disease modification: altering the rate of somatic expansion of the HTT CAG repeat or altering the resulting CAG threshold length-triggered toxicity process. Here, we evaluated the mouse orthologs of two HD age-at-onset modifier genes, FAN1 and RRM2B, for an influence on somatic instability of the expanded CAG repeat in Htt CAG knock-in mice. Fan1 knock-out increased somatic expansion of Htt CAG repeats, in the juvenile- and the adult-onset HD ranges, whereas knock-out of Rrm2b did not greatly alter somatic Htt CAG repeat instability. Simultaneous knock-out of Mlh1, the ortholog of a third HD age-at-onset modifier gene (MLH1), which suppresses somatic expansion of the Htt knock-in CAG repeat, blocked the Fan1 knock-out-induced acceleration of somatic CAG expansion. This genetic interaction indicates that functional MLH1 is required for the CAG repeat destabilizing effect of FAN1 loss. Thus, in HD, it is uncertain whether the RRM2B modifier effect on timing of onset may be due to a DNA instability mechanism. In contrast, the FAN1 modifier effects reveal that functional FAN1 acts to suppress somatic CAG repeat expansion, likely in genetic interaction with other DNA instability modifiers whose combined effects can hasten or delay onset and other CAG repeat length-driven phenotypes.


Subject(s)
Cell Cycle Proteins/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Multifunctional Enzymes/genetics , MutL Protein Homolog 1/genetics , Ribonucleotide Reductases/genetics , Age of Onset , Animals , Disease Models, Animal , Genes, Modifier/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Huntington Disease/pathology , Mice , Mice, Knockout , Phenotype , Trinucleotide Repeat Expansion/genetics
6.
Hum Mol Genet ; 29(15): 2551-2567, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32761094

ABSTRACT

The expanded HTT CAG repeat causing Huntington's disease (HD) exhibits somatic expansion proposed to drive the rate of disease onset by eliciting a pathological process that ultimately claims vulnerable cells. To gain insight into somatic expansion in humans, we performed comprehensive quantitative analyses of CAG expansion in ~50 central nervous system (CNS) and peripheral postmortem tissues from seven adult-onset and one juvenile-onset HD individual. We also assessed ATXN1 CAG repeat expansion in brain regions of an individual with a neurologically and pathologically distinct repeat expansion disorder, spinocerebellar ataxia type 1 (SCA1). Our findings reveal similar profiles of tissue instability in all HD individuals, which, notably, were also apparent in the SCA1 individual. CAG expansion was observed in all tissues, but to different degrees, with multiple cortical regions and neostriatum tending to have the greatest instability in the CNS, and liver in the periphery. These patterns indicate different propensities for CAG expansion contributed by disease locus-independent trans-factors and demonstrate that expansion per se is not sufficient to cause cell type or disease-specific pathology. Rather, pathology may reflect distinct toxic processes triggered by different repeat lengths across cell types and diseases. We also find that the HTT CAG length-dependent expansion propensity of an individual is reflected in all tissues and in cerebrospinal fluid. Our data indicate that peripheral cells may be a useful source to measure CAG expansion in biomarker assays for therapeutic efforts, prompting efforts to dissect underlying mechanisms of expansion that may differ between the brain and periphery.


Subject(s)
Huntington Disease/genetics , Spinocerebellar Ataxias/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics , Adult , Aged , Autopsy , Central Nervous System/pathology , Child , Female , Humans , Huntingtin Protein/genetics , Huntington Disease/diagnostic imaging , Huntington Disease/pathology , Male , Middle Aged , Neostriatum/diagnostic imaging , Neostriatum/metabolism , Neostriatum/pathology , Spinocerebellar Ataxias/diagnostic imaging , Spinocerebellar Ataxias/pathology
7.
Am J Hum Genet ; 103(3): 349-357, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30122542

ABSTRACT

Age at onset of Huntington disease, an inherited neurodegenerative disorder, is influenced by the size of the disease-causing CAG trinucleotide repeat expansion in HTT and by genetic modifier loci on chromosomes 8 and 15. Stratifying by modifier genotype, we have examined putamen volume, total motor score (TMS), and symbol digit modalities test (SDMT) scores, both at study entry and longitudinally, in normal controls and CAG-expansion carriers who were enrolled prior to the emergence of manifest HD in the PREDICT-HD study. The modifiers, which included onset-hastening and onset-delaying alleles on chromosome 15 and an onset-hastening allele on chromosome 8, revealed no major effect in controls but distinct patterns of modification in prediagnosis HD subjects. Putamen volume at study entry showed evidence of reciprocal modification by the chromosome 15 alleles, but the rate of loss of putamen volume was modified only by the deleterious chromosome 15 allele. By contrast, both alleles modified the rate of change of the SDMT score, but neither had an effect on the TMS. The influence of the chromosome 8 modifier was evident only in the rate of TMS increase. The data indicate that (1) modification of pathogenesis can occur early in the prediagnosis phase, (2) the modifier loci act in genetic interaction with the HD mutation rather than through independent additive effects, and (3) HD subclinical phenotypes are differentially influenced by each modifier, implying distinct effects in different cells or tissues. Together, these findings indicate the potential benefit of using genetic modifier strategies for dissecting the prediagnosis pathogenic process in HD.


Subject(s)
Huntington Disease/genetics , Mutation/genetics , Adult , Alleles , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 8/genetics , Female , Genotype , Humans , Huntingtin Protein/genetics , Male , Phenotype , Trinucleotide Repeat Expansion/genetics
8.
Hum Mol Genet ; 26(19): 3859-3867, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28934397

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disease caused by an expanded CAG repeat in HTT. Many clinical characteristics of HD such as age at motor onset are determined largely by the size of HTT CAG repeat. However, emerging evidence strongly supports a role for other genetic factors in modifying the disease pathogenesis driven by mutant huntingtin. A recent genome-wide association analysis to discover genetic modifiers of HD onset age provided initial evidence for modifier loci on chromosomes 8 and 15 and suggestive evidence for a locus on chromosome 3. Here, genotyping of candidate single nucleotide polymorphisms in a cohort of 3,314 additional HD subjects yields independent confirmation of the former two loci and moves the third to genome-wide significance at MLH1, a locus whose mouse orthologue modifies CAG length-dependent phenotypes in a Htt-knock-in mouse model of HD. Both quantitative and dichotomous association analyses implicate a functional variant on ∼32% of chromosomes with the beneficial modifier effect that delays HD motor onset by 0.7 years/allele. Genomic DNA capture and sequencing of a modifier haplotype localize the functional variation to a 78 kb region spanning the 3'end of MLH1 and the 5'end of the neighboring LRRFIP2, and marked by an isoleucine-valine missense variant in MLH1. Analysis of expression Quantitative Trait Loci (eQTLs) provides modest support for altered regulation of MLH1 and LRRFIP2, raising the possibility that the modifier affects regulation of both genes. Finally, polygenic modification score and heritability analyses suggest the existence of additional genetic modifiers, supporting expanded, comprehensive genetic analysis of larger HD datasets.


Subject(s)
Huntingtin Protein/genetics , MutL Protein Homolog 1/genetics , Alleles , Animals , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 8 , Disease Models, Animal , Genes, Modifier/genetics , Genome-Wide Association Study , Genotype , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , MutL Protein Homolog 1/metabolism , Phenotype , Polymorphism, Single Nucleotide/genetics , Trinucleotide Repeats
9.
Hum Mol Genet ; 26(7): 1258-1267, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28165127

ABSTRACT

Huntington's disease (HD) reflects dominant consequences of a CAG repeat expansion mutation in HTT. Expanded CAG repeat size is the primary determinant of age at onset and age at death in HD. Although HD pathogenesis is driven by the expanded CAG repeat, whether the mutation influences the expression levels of mRNA and protein from the disease allele is not clear due to the lack of sensitive allele-specific quantification methods and the presence of confounding factors. To determine the impact of CAG expansion at the molecular level, we have developed novel allele-specific HTT mRNA and protein quantification methods based on principles of multiplex ligation-dependent probe amplification and targeted MS/MS parallel reaction monitoring, respectively. These assays, exhibiting high levels of specificity and sensitivity, were designed to distinguish allelic products based upon expressed polymorphic variants in HTT, including rs149 109 767. To control for other cis-haplotype variations, we applied allele-specific quantification assays to a panel of HD lymphoblastoid cell lines, each carrying the major European disease haplotype (i.e. hap.01) on the mutant chromosome. We found that steady state levels of HTT mRNA and protein were not associated with expanded CAG repeat length. Rather, the products of mutant and normal alleles, both mRNA and protein, were balanced, thereby arguing that a cis-regulatory effect of the expanded CAG repeat is not a critical component of the underlying mechanism of HD. These robust allele-specific assays could prove valuable for monitoring the impact of allele-specific gene silencing strategies currently being explored as therapeutic interventions in HD.


Subject(s)
Brain/metabolism , Huntingtin Protein/biosynthesis , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Adult , Age of Onset , Alleles , Autopsy , Brain/pathology , Female , Gene Expression Regulation , Humans , Huntingtin Protein/genetics , Huntington Disease/pathology , Male , RNA, Messenger/biosynthesis
10.
Hum Mol Genet ; 26(5): 913-922, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28334820

ABSTRACT

Huntington's disease is a dominantly inherited neurodegenerative disease caused by the expansion of a CAG repeat in the HTT gene. In addition to the length of the CAG expansion, factors such as genetic background have been shown to contribute to the age at onset of neurological symptoms. A central challenge in understanding the disease progression that leads from the HD mutation to massive cell death in the striatum is the ability to characterize the subtle and early functional consequences of the CAG expansion longitudinally. We used dense time course sampling between 4 and 20 postnatal weeks to characterize early transcriptomic, molecular and cellular phenotypes in the striatum of six distinct knock-in mouse models of the HD mutation. We studied the effects of the HttQ111 allele on the C57BL/6J, CD-1, FVB/NCr1, and 129S2/SvPasCrl genetic backgrounds, and of two additional alleles, HttQ92 and HttQ50, on the C57BL/6J background. We describe the emergence of a transcriptomic signature in HttQ111/+ mice involving hundreds of differentially expressed genes and changes in diverse molecular pathways. We also show that this time course spanned the onset of mutant huntingtin nuclear localization phenotypes and somatic CAG-length instability in the striatum. Genetic background strongly influenced the magnitude and age at onset of these effects. This work provides a foundation for understanding the earliest transcriptional and molecular changes contributing to HD pathogenesis.


Subject(s)
Corpus Striatum/metabolism , Huntingtin Protein/genetics , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Corpus Striatum/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genetic Background , Genomic Instability/genetics , Humans , Huntingtin Protein/biosynthesis , Huntington Disease/pathology , Mice , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Phenotype , Transcriptome/genetics
11.
Am J Hum Genet ; 98(2): 287-98, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26849111

ABSTRACT

Huntington disease (HD) is caused by an expanded HTT CAG repeat that leads in a length-dependent, completely dominant manner to onset of a characteristic movement disorder. HD also displays early mortality, so we tested whether the expanded CAG repeat exerts a dominant influence on age at death and on the duration of clinical disease. We found that, as with clinical onset, HD age at death is determined by expanded CAG-repeat length and has no contribution from the normal CAG allele. Surprisingly, disease duration is independent of the mutation's length. It is also unaffected by a strong genetic modifier of HD motor onset. These findings suggest two parsimonious alternatives. (1) HD pathogenesis is driven by mutant huntingtin, but before or near motor onset, sufficient CAG-driven damage occurs to permit CAG-independent processes and then lead to eventual death. In this scenario, some pathological changes and their clinical correlates could still worsen in a CAG-driven manner after disease onset, but these CAG-related progressive changes do not themselves determine duration. Alternatively, (2) HD pathogenesis is driven by mutant huntingtin acting in a CAG-dependent manner with different time courses in multiple cell types, and the cellular targets that lead to motor onset and death are different and independent. In this scenario, processes driven by HTT CAG length lead directly to death but not via the striatal pathology associated with motor manifestations. Each scenario has important ramifications for the design and testing of potential therapeutics, especially those aimed at preventing or delaying characteristic motor manifestations.


Subject(s)
Huntington Disease/genetics , Mutation , Nerve Tissue Proteins/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Alleles , Child , Child, Preschool , Cohort Studies , Corpus Striatum/metabolism , Haplotypes , Humans , Huntingtin Protein , Huntington Disease/mortality , Middle Aged , Nerve Tissue Proteins/metabolism , Young Adult
12.
Hum Mol Genet ; 25(20): 4566-4576, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28172889

ABSTRACT

A comprehensive genetics-based precision medicine strategy to selectively and permanently inactivate only mutant, not normal allele, could benefit many dominantly inherited disorders. Here, we demonstrate the power of our novel strategy of inactivating the mutant allele using haplotype-specific CRISPR/Cas9 target sites in Huntington's disease (HD), a late-onset neurodegenerative disorder due to a toxic dominant gain-of-function CAG expansion mutation. Focusing on improving allele specificity, we combined extensive knowledge of huntingtin (HTT) gene haplotype structure with a novel personalized allele-selective CRISPR/Cas9 strategy based on Protospacer Adjacent Motif (PAM)-altering SNPs to target patient-specific CRISPR/Cas9 sites, aiming at the mutant HTT allele-specific inactivation for a given diplotype. As proof-of-principle, simultaneously using two CRISPR/Cas9 guide RNAs (gRNAs) that depend on PAM sites generated by SNP alleles on the mutant chromosome, we selectively excised ∼44 kb DNA spanning promoter region, transcription start site, and the CAG expansion mutation of the mutant HTT gene, resulting in complete inactivation of the mutant allele without impacting the normal allele. This excision on the disease chromosome completely prevented the generation of mutant HTT mRNA and protein, unequivocally indicating permanent mutant allele-specific inactivation of the HD mutant allele. The perfect allele selectivity with broad applicability of our strategy in disorders with diverse disease haplotypes should also support precision medicine through inactivation of many other gain-of-function mutations.


Subject(s)
CRISPR-Cas Systems , Genetic Therapy/methods , Huntingtin Protein/genetics , Huntington Disease/therapy , Trinucleotide Repeat Expansion , Cell Line , Fibroblasts/metabolism , Haplotypes , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Precision Medicine/methods
13.
Am J Hum Genet ; 97(3): 435-44, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26320893

ABSTRACT

Huntington disease (HD) reflects the dominant consequences of a CAG-repeat expansion in HTT. Analysis of common SNP-based haplotypes has revealed that most European HD subjects have distinguishable HTT haplotypes on their normal and disease chromosomes and that ∼50% of the latter share the same major HD haplotype. We reasoned that sequence-level investigation of this founder haplotype could provide significant insights into the history of HD and valuable information for gene-targeting approaches. Consequently, we performed whole-genome sequencing of HD and control subjects from four independent families in whom the major European HD haplotype segregates with the disease. Analysis of the full-sequence-based HTT haplotype indicated that these four families share a common ancestor sufficiently distant to have permitted the accumulation of family-specific variants. Confirmation of new CAG-expansion mutations on this haplotype suggests that unlike most founders of human disease, the common ancestor of HD-affected families with the major haplotype most likely did not have HD. Further, availability of the full sequence data validated the use of SNP imputation to predict the optimal variants for capturing heterozygosity in personalized allele-specific gene-silencing approaches. As few as ten SNPs are capable of revealing heterozygosity in more than 97% of European HD subjects. Extension of allele-specific silencing strategies to the few remaining homozygous individuals is likely to be achievable through additional known SNPs and discovery of private variants by complete sequencing of HTT. These data suggest that the current development of gene-based targeting for HD could be extended to personalized allele-specific approaches in essentially all HD individuals of European ancestry.


Subject(s)
Evolution, Molecular , Haplotypes/genetics , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Trinucleotide Repeat Expansion/genetics , White People/genetics , Base Sequence , Founder Effect , Heterozygote , Humans , Huntingtin Protein , Molecular Sequence Data , Pedigree , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
14.
Nature ; 466(7303): 243-7, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20555325

ABSTRACT

Sialic acid acetylesterase (SIAE) is an enzyme that negatively regulates B lymphocyte antigen receptor signalling and is required for the maintenance of immunological tolerance in mice. Heterozygous loss-of-function germline rare variants and a homozygous defective polymorphic variant of SIAE were identified in 24/923 subjects of European origin with relatively common autoimmune disorders and in 2/648 controls of European origin. All heterozygous loss-of-function SIAE mutations tested were capable of functioning in a dominant negative manner. A homozygous secretion-defective polymorphic variant of SIAE was catalytically active, lacked the ability to function in a dominant negative manner, and was seen in eight autoimmune subjects but in no control subjects. The odds ratio for inheriting defective SIAE alleles was 8.6 in all autoimmune subjects, 8.3 in subjects with rheumatoid arthritis, and 7.9 in subjects with type I diabetes. Functionally defective SIAE rare and polymorphic variants represent a strong genetic link to susceptibility in relatively common human autoimmune disorders.


Subject(s)
Acetylesterase/genetics , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmunity/genetics , Carboxylic Ester Hydrolases/genetics , Genetic Predisposition to Disease/genetics , Germ-Line Mutation/genetics , N-Acetylneuraminic Acid/metabolism , Acetylation , Acetylesterase/metabolism , Alleles , Animals , Antibodies, Antinuclear/blood , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , B-Lymphocytes/metabolism , Biocatalysis , Carboxylic Ester Hydrolases/metabolism , Case-Control Studies , Cell Line , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/genetics , Europe/ethnology , Exons/genetics , Humans , Mice , Odds Ratio , Polymorphism, Single Nucleotide/genetics , Sample Size , Sequence Analysis, DNA
15.
PLoS Genet ; 9(10): e1003930, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204323

ABSTRACT

The Huntington's disease gene (HTT) CAG repeat mutation undergoes somatic expansion that correlates with pathogenesis. Modifiers of somatic expansion may therefore provide routes for therapies targeting the underlying mutation, an approach that is likely applicable to other trinucleotide repeat diseases. Huntington's disease Hdh(Q111) mice exhibit higher levels of somatic HTT CAG expansion on a C57BL/6 genetic background (B6.Hdh(Q111) ) than on a 129 background (129.Hdh(Q111) ). Linkage mapping in (B6x129).Hdh(Q111) F2 intercross animals identified a single quantitative trait locus underlying the strain-specific difference in expansion in the striatum, implicating mismatch repair (MMR) gene Mlh1 as the most likely candidate modifier. Crossing B6.Hdh(Q111) mice onto an Mlh1 null background demonstrated that Mlh1 is essential for somatic CAG expansions and that it is an enhancer of nuclear huntingtin accumulation in striatal neurons. Hdh(Q111) somatic expansion was also abolished in mice deficient in the Mlh3 gene, implicating MutLγ (MLH1-MLH3) complex as a key driver of somatic expansion. Strikingly, Mlh1 and Mlh3 genes encoding MMR effector proteins were as critical to somatic expansion as Msh2 and Msh3 genes encoding DNA mismatch recognition complex MutSß (MSH2-MSH3). The Mlh1 locus is highly polymorphic between B6 and 129 strains. While we were unable to detect any difference in base-base mismatch or short slipped-repeat repair activity between B6 and 129 MLH1 variants, repair efficiency was MLH1 dose-dependent. MLH1 mRNA and protein levels were significantly decreased in 129 mice compared to B6 mice, consistent with a dose-sensitive MLH1-dependent DNA repair mechanism underlying the somatic expansion difference between these strains. Together, these data identify Mlh1 and Mlh3 as novel critical genetic modifiers of HTT CAG instability, point to Mlh1 genetic variation as the likely source of the instability difference in B6 and 129 strains and suggest that MLH1 protein levels play an important role in driving of the efficiency of somatic expansions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Huntington Disease/genetics , Nuclear Proteins/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Disease Models, Animal , Genetic Association Studies , Genome-Wide Association Study , Genomic Instability , Humans , Huntington Disease/pathology , Mice , MutL Protein Homolog 1 , MutL Proteins , RNA, Messenger
16.
Hum Mol Genet ; 22(16): 3227-38, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23595883

ABSTRACT

In Huntington's disease (HD), the size of the expanded HTT CAG repeat mutation is the primary driver of the processes that determine age at onset of motor symptoms. However, correlation of cellular biochemical parameters also extends across the normal repeat range, supporting the view that the CAG repeat represents a functional polymorphism with dominant effects determined by the longer allele. A central challenge to defining the functional consequences of this single polymorphism is the difficulty of distinguishing its subtle effects from the multitude of other sources of biological variation. We demonstrate that an analytical approach based upon continuous correlation with CAG size was able to capture the modest (∼21%) contribution of the repeat to the variation in genome-wide gene expression in 107 lymphoblastoid cell lines, with alleles ranging from 15 to 92 CAGs. Furthermore, a mathematical model from an iterative strategy yielded predicted CAG repeat lengths that were significantly positively correlated with true CAG allele size and negatively correlated with age at onset of motor symptoms. Genes negatively correlated with repeat size were also enriched in a set of genes whose expression were CAG-correlated in human HD cerebellum. These findings both reveal the relatively small, but detectable impact of variation in the CAG allele in global data in these peripheral cells and provide a strategy for building multi-dimensional data-driven models of the biological network that drives the HD disease process by continuous analysis across allelic panels of neuronal cells vulnerable to the dominant effects of the HTT CAG repeat.


Subject(s)
Gene Expression , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Trinucleotide Repeats/genetics , Age of Onset , Alleles , Cell Line , Cerebellum/metabolism , Female , Gene Expression Regulation , Humans , Huntingtin Protein , Huntington Disease/diagnosis , Huntington Disease/metabolism , Male , Models, Genetic , Polymorphism, Genetic , Reproducibility of Results , Transcriptome
17.
Am J Hum Genet ; 90(3): 434-44, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22387017

ABSTRACT

Age at the onset of motor symptoms in Huntington disease (HD) is determined largely by the length of a CAG repeat expansion in HTT but is also influenced by other genetic factors. We tested whether common genetic variation near the mutation site is associated with differences in the distribution of expanded CAG alleles or age at the onset of motor symptoms. To define disease-associated single-nucleotide polymorphisms (SNPs), we compared 4p16.3 SNPs in HD subjects with population controls in a case:control strategy, which revealed that the strongest signals occurred at a great distance from the HD mutation as a result of "synthetic association" with SNP alleles that are of low frequency in population controls. Detailed analysis delineated a prominent ancestral haplotype that accounted for ∼50% of HD chromosomes and extended to at least 938 kb on about half of these. Together, the seven most abundant haplotypes accounted for ∼83% of HD chromosomes. Neither the extended shared haplotype nor the individual local HTT haplotypes were associated with altered CAG-repeat length distribution or residual age at the onset of motor symptoms, arguing against modification of these disease features by common cis-regulatory elements. Similarly, the 11 most frequent control haplotypes showed no trans-modifier effect on age at the onset of motor symptoms. Our results argue against common local regulatory variation as a factor influencing HD pathogenesis, suggesting that genetic modifiers be sought elsewhere in the genome. They also indicate that genome-wide association analysis with a small number of cases can be effective for regional localization of genetic defects, even when a founder effect accounts for only a fraction of the disorder.


Subject(s)
Chromosomes, Human, Pair 4 , Huntington Disease/genetics , Age of Onset , Alleles , Case-Control Studies , Founder Effect , Genome-Wide Association Study/methods , Haplotypes , Humans , Mutation , Polymorphism, Single Nucleotide , Trinucleotide Repeats
18.
Mamm Genome ; 26(3-4): 119-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645993

ABSTRACT

Huntington's disease (HD) is a dominant neurodegenerative disorder that is due to expansion of an unstable HTT CAG repeat for which genome-wide genetic scans are now revealing chromosome regions that contain disease-modifying genes. We have explored a novel human-mouse cross-species functional prioritisation approach, by evaluating the HD modifier 6q23-24 linkage interval. This unbiased strategy employs C57BL/6J (B6J) Hdh(Q111) knock-in mice, replicates of the HD mutation, and the C57BL/6J-chr10(A/J)/NaJ chromosome substitution strain (CSS10), in which only chromosome 10 (chr10), in synteny with the human 6q23-24 region, is derived from the A/J (AJ) strain. Crosses were performed to assess the possibility of dominantly acting chr10 AJ-B6J variants of strong effect that may modulate CAG-dependent Hdh(Q111/+) phenotypes. Testing of F1 progeny confirmed that a single AJ chromosome had a significant effect on the rate of body weight gain and in Hdh(Q111) mice the AJ chromosome was associated subtle alterations in somatic CAG instability in the liver and the formation of intra-nuclear inclusions, as well as DARPP-32 levels, in the striatum. These findings in relatively small cohorts are suggestive of dominant chr10 AJ-B6 variants that may modify effects of the CAG expansion, and encourage a larger study with CSS10 and sub-strains. This cross-species approach may therefore be suited to functional in vivo prioritisation of genomic regions harbouring genes that can modify the early effects of the HD mutation.


Subject(s)
Chromosomes, Mammalian , Crosses, Genetic , Huntington Disease/genetics , Quantitative Trait Loci , Alleles , Animals , Body Weight , Chromosomes, Human , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Female , Gene Knock-In Techniques , Genetic Variation , Genomic Instability , Genotype , Humans , Huntingtin Protein , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phenotype , Trinucleotide Repeats
19.
Bipolar Disord ; 17(4): 403-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25726852

ABSTRACT

OBJECTIVES: Huntington's disease is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric symptoms that are caused by huntingtin gene (HTT) CAG trinucleotide repeat alleles of 36 or more units. A greater than expected prevalence of incompletely penetrant HTT CAG repeat alleles observed among individuals diagnosed with major depressive disorder raises the possibility that another mood disorder, bipolar disorder, could likewise be associated with Huntington's disease. METHODS: We assessed the distribution of HTT CAG repeat alleles in a cohort of individuals with bipolar disorder. HTT CAG allele sizes from 2,229 Caucasian individuals diagnosed with DSM-IV bipolar disorder were compared to allele sizes in 1,828 control individuals from multiple cohorts. RESULTS: We found that HTT CAG repeat alleles > 35 units were observed in only one of 4,458 chromosomes from individuals with bipolar disorder, compared to three of 3,656 chromosomes from control subjects. CONCLUSIONS: These findings do not support an association between bipolar disorder and Huntington's disease.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Trinucleotide Repeats/genetics , Adult , Alleles , Amyotrophic Lateral Sclerosis/genetics , Bipolar Disorder/genetics , Depressive Disorder, Major/genetics , Female , Genotype , Humans , Huntingtin Protein , Huntington Disease/diagnosis , Male , Middle Aged , Penetrance , Prevalence , Statistics as Topic
20.
Am J Med Genet B Neuropsychiatr Genet ; 168B(2): 135-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25656686

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder characterized by involuntary choreic movements, cognitive impairment, and behavioral changes, caused by the expansion of an unstable CAG repeat in HTT. We characterized the genetic diversity of the HD mutation by performing an extensive haplotype analysis of ∼1Mb region flanking HTT in over 300 HD families of Portuguese origin. We observed that haplotype A, marked by HTT delta2642, was enriched in HD chromosomes and carried the two largest expansions reported in the Portuguese population. However, the most frequent HD haplotype B carried one of the largest (+12 CAGs) expansions, which resulted in an allele class change to full penetrance. Despite having a normal CAG distribution skewed to the higher end of the range, these two core haplotypes had similar expanded CAG repeat sizes compared to the other major core haplotypes (C and D) and there was no statistical difference in transmitted repeat instability across haplotypes. We observed a diversity of HTT region haplotypes in both normal and expanded chromosomes, representative of more than one ancestral chromosome underlying HD in Portugal, where multiple independent events on distinct chromosome 4 haplotypes have given rise to expansion into the pathogenic range.


Subject(s)
Chromosomes, Human, Pair 4/genetics , Haplotypes/genetics , Huntington Disease/genetics , Alleles , Base Pairing/genetics , Family , Female , Genomic Instability , Humans , Male , Pedigree , Polymorphism, Single Nucleotide/genetics , Portugal , Trinucleotide Repeat Expansion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL