107 results on '"Seung Kwak"'
Search Results
2. A genetic association study of glutamine-encoding DNA sequence structures, somatic CAG expansion, and DNA repair gene variants, with Huntington disease clinical outcomes
- Author
-
Marc Ciosi, Alastair Maxwell, Sarah A. Cumming, Davina J. Hensman Moss, Asma M. Alshammari, Michael D. Flower, Alexandra Durr, Blair R. Leavitt, Raymund A.C. Roos, Peter Holmans, Lesley Jones, Douglas R. Langbehn, Seung Kwak, Sarah J. Tabrizi, and Darren G. Monckton
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Huntington disease (HD) is caused by an unstable CAG/CAA repeat expansion encoding a toxic polyglutamine tract. Here, we tested the hypotheses that HD outcomes are impacted by somatic expansion of, and polymorphisms within, the HTT CAG/CAA glutamine-encoding repeat, and DNA repair genes. Methods: The sequence of the glutamine-encoding repeat and the proportion of somatic CAG expansions in blood DNA from participants inheriting 40 to 50 CAG repeats within the TRACK-HD and Enroll-HD cohorts were determined using high-throughput ultra-deep-sequencing. Candidate gene polymorphisms were genotyped using kompetitive allele-specific PCR (KASP). Genotypic associations were assessed using time-to-event and regression analyses. Findings: Using data from 203 TRACK-HD and 531 Enroll-HD participants, we show that individuals with higher blood DNA somatic CAG repeat expansion scores have worse HD outcomes: a one-unit increase in somatic expansion score was associated with a Cox hazard ratio for motor onset of 3·05 (95% CI = 1·94 to 4·80, p = 1·3 × 10−6). We also show that individual-specific somatic expansion scores are associated with variants in FAN1 (pFDR = 4·8 × 10-6), MLH3 (pFDR = 8·0 × 10−4), MLH1 (pFDR = 0·004) and MSH3 (pFDR = 0·009). We also show that HD outcomes are best predicted by the number of pure CAGs rather than total encoded-glutamines. Interpretation: These data establish pure CAG length, rather than encoded-glutamine, as the key inherited determinant of downstream pathophysiology. These findings have implications for HD diagnostics, and support somatic expansion as a mechanistic link for genetic modifiers of clinical outcomes, a driver of disease, and potential therapeutic target in HD and related repeat expansion disorders. Funding: CHDI Foundation. Keywords: Genetic association study, Somatic expansion, DNA repair, Huntington disease
- Published
- 2019
- Full Text
- View/download PDF
3. Novel DNA Aptamers that Bind to Mutant Huntingtin and Modify Its Activity
- Author
-
Baehyun Shin, Roy Jung, Hyejin Oh, Gwen E. Owens, Hyeongseok Lee, Seung Kwak, Ramee Lee, Susan L. Cotman, Jong-Min Lee, Marcy E. MacDonald, Ji-Joon Song, Ravi Vijayvargia, and Ihn Sik Seong
- Subjects
Huntington's disease ,full-length huntingtin ,single-stranded oligonucleotide ,HEAT repeats ,polycomb repressive complex 2 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The CAG repeat expansion that elongates the polyglutamine tract in huntingtin is the root genetic cause of Huntington’s disease (HD), a debilitating neurodegenerative disorder. This seemingly slight change to the primary amino acid sequence alters the physical structure of the mutant protein and alters its activity. We have identified a set of G-quadruplex-forming DNA aptamers (MS1, MS2, MS3, MS4) that bind mutant huntingtin proximal to lysines K2932/K2934 in the C-terminal CTD-II domain. Aptamer binding to mutant huntingtin abrogated the enhanced polycomb repressive complex 2 (PRC2) stimulatory activity conferred by the expanded polyglutamine tract. In HD, but not normal, neuronal progenitor cells (NPCs), MS3 aptamer co-localized with endogenous mutant huntingtin and was associated with significantly decreased PRC2 activity. Furthermore, MS3 transfection protected HD NPCs against starvation-dependent stress with increased ATP. Therefore, DNA aptamers can preferentially target mutant huntingtin and modulate a gain of function endowed by the elongated polyglutamine segment. These mutant huntingtin binding aptamers provide novel molecular tools for delineating the effects of the HD mutation and encourage mutant huntingtin structure-based approaches to therapeutic development.
- Published
- 2018
- Full Text
- View/download PDF
4. Early and brain region-specific decrease of de novo cholesterol biosynthesis in Huntington's disease: A cross-validation study in Q175 knock-in mice
- Author
-
Mahalakshmi Shankaran, Eleonora Di Paolo, Valerio Leoni, Claudio Caccia, Costanza Ferrari Bardile, Hussein Mohammed, Stefano Di Donato, Seung Kwak, Deanna Marchionini, Scott Turner, Elena Cattaneo, and Marta Valenza
- Subjects
Huntington ,Brain cholesterol ,Lathosterol ,Striatum ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Cholesterol precursors and cholesterol levels are reduced in brain regions of Huntington's disease (HD) mice. Here we quantified the rate of in vivo de novo cholesterol biosynthesis in the HD brain. Samples from different brain regions and blood of the heterozygous knock-in mouse model carrying 175 CAG repeats (Q175) at different phenotypic stages were processed independently by two research units to quantify cholesterol synthesis rate by 2H2O labeling and measure the concentrations of lathosterol, cholesterol and its brain-specific cholesterol catabolite 24-hydroxy-cholesterol (24OHC) by isotope dilution mass spectrometry. The daily synthesis rate of cholesterol and the corresponding concentration of lathosterol were significantly reduced in the striatum of heterozygous Q175 mice early in the disease course. We also report that the decrease in lathosterol was inversely correlated with CAG-size at symptomatic stage, as observed in striatal samples from an allelic series of HD mice. There was also a significant correlation between the fractional synthesis rates of total cholesterol and 24OHC in brain of wild-type (WT) and Q175 mice, supporting the evidence that plasma 24OHC may reflect cholesterol synthesis in the adult brain. This comprehensive analysis demonstrates consistent cholesterol biosynthesis defects in HD mouse models and suggests that plasma 24OHC may serve as a biomarker of brain cholesterol metabolism.
- Published
- 2017
- Full Text
- View/download PDF
5. Population-specific genetic modification of Huntington's disease in Venezuela.
- Author
-
Michael J Chao, Kyung-Hee Kim, Jun Wan Shin, Diane Lucente, Vanessa C Wheeler, Hong Li, Jared C Roach, Leroy Hood, Nancy S Wexler, Laura B Jardim, Peter Holmans, Lesley Jones, Michael Orth, Seung Kwak, Marcy E MacDonald, James F Gusella, and Jong-Min Lee
- Subjects
Genetics ,QH426-470 - Abstract
Modifiers of Mendelian disorders can provide insights into disease mechanisms and guide therapeutic strategies. A recent genome-wide association (GWA) study discovered genetic modifiers of Huntington's disease (HD) onset in Europeans. Here, we performed whole genome sequencing and GWA analysis of a Venezuelan HD cluster whose families were crucial for the original mapping of the HD gene defect. The Venezuelan HD subjects develop motor symptoms earlier than their European counterparts, implying the potential for population-specific modifiers. The main Venezuelan HD family inherits HTT haplotype hap.03, which differs subtly at the sequence level from European HD hap.03, suggesting a different ancestral origin but not explaining the earlier age at onset in these Venezuelans. GWA analysis of the Venezuelan HD cluster suggests both population-specific and population-shared genetic modifiers. Genome-wide significant signals at 7p21.2-21.1 and suggestive association signals at 4p14 and 17q21.2 are evident only in Venezuelan HD, but genome-wide significant association signals at the established European chromosome 15 modifier locus are improved when Venezuelan HD data are included in the meta-analysis. Venezuelan-specific association signals on chromosome 7 center on SOSTDC1, which encodes a bone morphogenetic protein antagonist. The corresponding SNPs are associated with reduced expression of SOSTDC1 in non-Venezuelan tissue samples, suggesting that interaction of reduced SOSTDC1 expression with a population-specific genetic or environmental factor may be responsible for modification of HD onset in Venezuela. Detection of population-specific modification in Venezuelan HD supports the value of distinct disease populations in revealing novel aspects of a disease and population-relevant therapeutic strategies.
- Published
- 2018
- Full Text
- View/download PDF
6. MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice.
- Author
-
Peter Langfelder, Fuying Gao, Nan Wang, David Howland, Seung Kwak, Thomas F Vogt, Jeffrey S Aaronson, Jim Rosinski, Giovanni Coppola, Steve Horvath, and X William Yang
- Subjects
Medicine ,Science - Abstract
In Huntington's disease (HD) patients and in model organisms, messenger RNA transcriptome has been extensively studied; in contrast, comparatively little is known about expression and potential role of microRNAs. Using RNA-sequencing, we have quantified microRNA expression in four brain regions and liver, at three different ages, from an allelic series of HD model mice with increasing CAG length in the endogenous Huntingtin gene. Our analyses reveal CAG length-dependent microRNA expression changes in brain, with 159 microRNAs selectively altered in striatum, 102 in cerebellum, 51 in hippocampus, and 45 in cortex. In contrast, a progressive CAG length-dependent microRNA dysregulation was not observed in liver. We further identify microRNAs whose transcriptomic response to CAG length expansion differs significantly among the brain regions and validate our findings in data from a second, independent cohort of mice. Using existing mRNA expression data from the same animals, we assess the possible relationships between microRNA and mRNA expression and highlight candidate microRNAs that are negatively correlated with, and whose predicted targets are enriched in, CAG-length dependent mRNA modules. Several of our top microRNAs (Mir212/Mir132, Mir218, Mir128 and others) have been previously associated with aspects of neuronal development and survival. This study provides an extensive resource for CAG length-dependent changes in microRNA expression in disease-vulnerable and -resistant brain regions in HD mice, and provides new insights for further investigation of microRNAs in HD pathogenesis and therapeutics.
- Published
- 2018
- Full Text
- View/download PDF
7. HdhQ111 Mice Exhibit Tissue Specific Metabolite Profiles that Include Striatal Lipid Accumulation.
- Author
-
Jeffrey B Carroll, Amy Deik, Elisa Fossale, Rory M Weston, Jolene R Guide, Jamshid Arjomand, Seung Kwak, Clary B Clish, and Marcy E MacDonald
- Subjects
Medicine ,Science - Abstract
The HTT CAG expansion mutation causes Huntington's Disease and is associated with a wide range of cellular consequences, including altered metabolism. The mutant allele is expressed widely, in all tissues, but the striatum and cortex are especially vulnerable to its effects. To more fully understand this tissue-specificity, early in the disease process, we asked whether the metabolic impact of the mutant CAG expanded allele in heterozygous B6.HdhQ111/+ mice would be common across tissues, or whether tissues would have tissue-specific responses and whether such changes may be affected by diet. Specifically, we cross-sectionally examined steady state metabolite concentrations from a range of tissues (plasma, brown adipose tissue, cerebellum, striatum, liver, white adipose tissue), using an established liquid chromatography-mass spectrometry pipeline, from cohorts of 8 month old mutant and wild-type littermate mice that were fed one of two different high-fat diets. The differential response to diet highlighted a proportion of metabolites in all tissues, ranging from 3% (7/219) in the striatum to 12% (25/212) in white adipose tissue. By contrast, the mutant CAG-expanded allele primarily affected brain metabolites, with 14% (30/219) of metabolites significantly altered, compared to wild-type, in striatum and 11% (25/224) in the cerebellum. In general, diet and the CAG-expanded allele both elicited metabolite changes that were predominantly tissue-specific and non-overlapping, with evidence for mutation-by-diet interaction in peripheral tissues most affected by diet. Machine-learning approaches highlighted the accumulation of diverse lipid species as the most genotype-predictive metabolite changes in the striatum. Validation experiments in cell culture demonstrated that lipid accumulation was also a defining feature of mutant HdhQ111 striatal progenitor cells. Thus, metabolite-level responses to the CAG expansion mutation in vivo were tissue specific and most evident in brain, where the striatum featured signature accumulation of a set of lipids including sphingomyelin, phosphatidylcholine, cholesterol ester and triglyceride species. Importantly, in the presence of the CAG mutation, metabolite changes were unmasked in peripheral tissues by an interaction with dietary fat, implying that the design of studies to discover metabolic changes in HD mutation carriers should include metabolic perturbations.
- Published
- 2015
- Full Text
- View/download PDF
8. Characterization of HTT inclusion size, location, and timing in the zQ175 mouse model of Huntington's disease: an in vivo high-content imaging study.
- Author
-
Nikisha Carty, Nadège Berson, Karsten Tillack, Christina Thiede, Diana Scholz, Karsten Kottig, Yalda Sedaghat, Christina Gabrysiak, George Yohrling, Heinz von der Kammer, Andreas Ebneth, Volker Mack, Ignacio Munoz-Sanjuan, and Seung Kwak
- Subjects
Medicine ,Science - Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Major pathological hallmarks of HD include inclusions of mutant huntingtin (mHTT) protein, loss of neurons predominantly in the caudate nucleus, and atrophy of multiple brain regions. However, the early sequence of histological events that manifest in region- and cell-specific manner has not been well characterized. Here we use a high-content histological approach to precisely monitor changes in HTT expression and characterize deposition dynamics of mHTT protein inclusion bodies in the recently characterized zQ175 knock-in mouse line. We carried out an automated multi-parameter quantitative analysis of individual cortical and striatal cells in tissue slices from mice aged 2-12 months and confirmed biochemical reports of an age-associated increase in mHTT inclusions in this model. We also found distinct regional and subregional dynamics for inclusion number, size and distribution with subcellular resolution. We used viral-mediated suppression of total HTT in the striatum of zQ175 mice as an example of a therapeutically-relevant but heterogeneously transducing strategy to demonstrate successful application of this platform to quantitatively assess target engagement and outcome on a cellular basis.
- Published
- 2015
- Full Text
- View/download PDF
9. Myostatin inhibition slows muscle atrophy in rodent models of amyotrophic lateral sclerosis
- Author
-
Erika L.F. Holzbaur, David S. Howland, Nicholas Weber, Karen Wallace, Yijin She, Seung Kwak, Lioudmilla A. Tchistiakova, Erin Murphy, Joseph Hinson, Riyez Karim, Xiang Yang Tan, Pamela Kelley, Kevin C. McGill, Gareth Williams, Carl Hobbs, Patrick Doherty, Margaret M. Zaleska, Menelas N. Pangalos, and Frank S. Walsh
- Subjects
Amyotrophic lateral sclerosis ,SOD1 ,Myostatin ,GDF-8 ,Motor neuron disease ,Motor neuron degeneration ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease leading to motor neuron cell death, but recent studies suggest that non-neuronal cells may contribute to the pathological mechanisms involved. Myostatin is a negative regulator of muscle growth whose function can be inhibited using neutralizing antibodies. In this study, we used transgenic mouse and rat models of ALS to test whether treatment with anti-myostatin antibody slows muscle atrophy, motor neuron loss, or disease onset and progression. Significant increases in muscle mass and strength were observed in myostatin-antibody-treated SOD1G93A mice and rats prior to disease onset and during early-stage disease. By late stage disease, only diaphragm muscle remained significantly different in treated animals in comparison to untreated controls. Myostatin inhibition did not delay disease onset nor extend survival in either the SOD1G93A mouse or rat. Together, these results indicate that inhibition of myostatin does not protect against the onset and progression of motor neuron degenerative disease. However, the preservation of skeletal muscle during early-stage disease and improved diaphragm morphology and function maintained through late stage disease suggest that anti-myostatin therapy may promote some improved muscle function in ALS.
- Published
- 2006
- Full Text
- View/download PDF
10. Posttranscriptional regulation of FAN1 by miR-124-3p at rs3512 underlies onset-delaying genetic modification in Huntington's disease.
- Author
-
Kyung-Hee Kim, Eun Pyo Hong, Yukyeong Lee, McLean, Zachariah L., Elezi, Emanuela, Lee, Ramee, Seung Kwak, McAllister, Branduff, Massey, Thomas H., Lobanov, Sergey, Holmans, Peter, Orth, Michael, Ciosi, Marc, Monckton, Darren G., Long, Jeffrey D., Lucente, Diane, Wheeler, Vanessa C., MacDonald, Marcy E., Gusella, James F., and Jong-Min Lee
- Subjects
HUNTINGTON disease ,LOCUS (Genetics) ,GENE expression ,GENETIC variation ,MESSENGER RNA - Abstract
Many Mendelian disorders, such as Huntington's disease (HD) and spinocerebellar ataxias, arise from expansions of CAG trinucleotide repeats. Despite the clear genetic causes, additional genetic factors may influence the rate of those monogenic disorders. Notably, genome-wide association studies discovered somewhat expected modifiers, particularly mismatch repair genes involved in the CAG repeat instability, impacting age at onset of HD. Strikingly, FAN1, previously unrelated to repeat instability, produced the strongest HD modification signals. Diverse FAN1 haplotypes independently modify HD, with rare genetic variants diminishing DNA binding or nuclease activity of the FAN1 protein, hastening HD onset. However, the mechanism behind the frequent and the most significant onset-delaying FAN1 haplotype lacking missense variations has remained elusive. Here, we illustrated that a microRNA acting on 3'-UTR (untranslated region) SNP rs3512, rather than transcriptional regulation, is responsible for the significant FAN1 expression quantitative trait loci signal and allelic imbalance in FAN1 messenger ribonucleic acid (mRNA), accounting for the most significant and frequent onset-delaying modifier haplotype in HD. Specifically, miR-124-3p selectively targets the reference allele at rs3512, diminishing the stability of FAN1 mRNA harboring that allele and consequently reducing its levels. Subsequent validation analyses, including the use of antagomir and 3'-UTR reporter vectors with swapped alleles, confirmed the specificity of miR-124-3p at rs3512. Together, these findings indicate that the alternative allele at rs3512 renders the FAN1 mRNA less susceptible to miR-124-3p-mediated posttranscriptional regulation, resulting in increased FAN1 levels and a subsequent delay in HD onset by mitigating CAG repeat instability. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Effects of nutritional status and support on the survival of COVID-19 patients in a single tertiary-center intensive care unit in Korea: a case-control study
- Author
-
Na Gyeong Lee, Hye Jin Kim, Ji Gyeong Kang, Do Hyeon Jeon, Mi Seung Kwak, and So Hyun Nam
- Published
- 2023
12. Huntingtin turnover: modulation of huntingtin degradation by cAMP-dependent protein kinase A (PKA) phosphorylation of C-HEAT domain Ser2550
- Author
-
Yejin Lee, Hyeongju Kim, Douglas Barker, Ravi Vijayvargia, Ranjit Singh Atwal, Harrison Specht, Hasmik Keshishian, Steven A Carr, Ramee Lee, Seung Kwak, Kyung-gi Hyun, Jacob Loupe, Marcy E MacDonald, Ji-Joon Song, and Ihn Sik Seong
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by an inherited unstable HTT CAG repeat that expands further, thereby eliciting a disease process that may be initiated by polyglutamine-expanded huntingtin or a short polyglutamine-product. Phosphorylation of selected candidate residues is reported to mediate polyglutamine-fragment degradation and toxicity. Here to support the discovery of phosphosites involved in the life-cycle of (full-length) huntingtin, we employed mass spectrometry-based phosphoproteomics to systematically identify sites in purified huntingtin and in the endogenous protein by proteomic and phosphoproteomic analyses of members of an HD neuronal progenitor cell panel. Our results bring total huntingtin phosphosites to 95, with more located in the N-HEAT domain relative to numbers in the Bridge and C-HEAT domains. Moreover, phosphorylation of C-HEAT Ser2550 by cAMP-dependent protein kinase (PKA), the top hit in kinase activity screens, was found to hasten huntingtin degradation, such that levels of the catalytic subunit (PRKACA) were inversely related to huntingtin levels. Taken together, these findings highlight categories of phosphosites that merit further study and provide a phosphosite kinase pair (pSer2550-PKA) with which to investigate the biological processes that regulate huntingtin degradation and thereby influence the steady state levels of huntingtin in HD cells.
- Published
- 2022
13. How do consumers’ perceptions of brands change?
- Author
-
Hee Seung Kwak, Jeong Ah Park, and Hyun-Hwa Lee
- Published
- 2022
14. How Does Monetary Policy Affect Prices of Corporate Loans?
- Author
-
Seung Kwak
- Abstract
This paper studies the impact of unanticipated monetary policy news around FOMC announcements on secondary market corporate loan spreads. I find that the reaction of loan spreads to monetary policy news is weaker than that of bond spreads: following an unanticipated monetary policy tightening (easing) shock, loan spreads do not increase (decrease) as much as bond spreads do. Decomposition of the spreads into compensations for expected defaults and risk premiums shows that differential reactions of loan and bond risk premiums are the main driver of the differential spread reactions. This paper further finds that the weaker loan spread reactions to monetary policy shocks are more pronounced for riskier loans. Lastly, reactions of primary market loan spreads to monetary policy shocks are also muted. These findings highlight heterogeneous impacts of monetary policy across different types of corporate credit markets, possibly reflecting heterogeneous investor demand responses to monetary policy in those markets.
- Published
- 2022
15. D01 Longitudinal analysis of somatic expansion of the HTT repeat in blood as a potential biomarker of somatic instability in Huntingon disease
- Author
-
Marc Ciosi, Robin Young, Seung Kwak, and Darren G Monckton
- Published
- 2022
16. Huntington’s Disease Pathogenesis: Two Sequential Components
- Author
-
Marcy E. MacDonald, Darren G. Monckton, Lesley Jones, Vanessa C. Wheeler, Michael Orth, Eun Pyo Hong, Seung Kwak, Peter Holmans, Jong-Min Lee, James F. Gusella, and Jeffrey D. Long
- Subjects
0301 basic medicine ,genetic association ,Locus (genetics) ,Disease ,Review ,Biology ,genotype-phenotype correlation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Huntington's disease ,medicine ,Humans ,genetics ,Cognitive decline ,Genetic Association Studies ,Genetic association ,Genetics ,modifier gene ,Huntingtin Protein ,Genes, Modifier ,medicine.disease ,Human genetics ,030104 developmental biology ,Huntington Disease ,Human genome ,Neurology (clinical) ,trinucleotide repeat expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery - Abstract
Historically, Huntington’s disease (HD; OMIM #143100) has played an important role in the enormous advances in human genetics seen over the past four decades. This familial neurodegenerative disorder involves variable onset followed by consistent worsening of characteristic abnormal movements along with cognitive decline and psychiatric disturbances. HD was the first autosomal disease for which the genetic defect was assigned to a position on the human chromosomes using only genetic linkage analysis with common DNA polymorphisms. This discovery set off a multitude of similar studies in other diseases, while the HD gene, later renamed HTT, and its vicinity in chromosome 4p16.3 then acted as a proving ground for development of technologies to clone and sequence genes based upon their genomic location, with the growing momentum of such advances fueling the Human Genome Project. The identification of the HD gene has not yet led to an effective treatment, but continued human genetic analysis of genotype-phenotype relationships in large HD subject populations, first at the HTT locus and subsequently genome-wide, has provided insights into pathogenesis that divide the course of the disease into two sequential, mechanistically distinct components.
- Published
- 2021
17. Approaches to Sequence the HTT CAG Repeat Expansion and Quantify Repeat Length Variation
- Author
-
Seung Kwak, Sarah A. Cumming, Darren G. Monckton, Ricardo Mouro Pinto, Graham Hamilton, A. Jennifer Morton, Eloise Larson, Vanessa C. Wheeler, William Tottey, Marc Ciosi, Afroditi Chatzi, Vilija Lomeikaite, Morton, Jennifer [0000-0003-0181-6346], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Research Report ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Somatic cell ,huntingtin ,repeat expansion ,Mice, Transgenic ,Biology ,Polymerase Chain Reaction ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Huntington's disease ,Genotype ,mental disorders ,medicine ,Animals ,Allele ,Sequence (medicine) ,Genetics ,Huntingtin Protein ,Massive parallel sequencing ,Mosaicism ,parallel sequencing ,Sequence Analysis, DNA ,Huntington disease ,medicine.disease ,Somatic mosaicism ,nervous system diseases ,030104 developmental biology ,Neurology (clinical) ,Trinucleotide repeat expansion ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery - Abstract
Background:\ud Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by the expansion of the HTT CAG repeat. Affected individuals inherit ≥36 repeats and longer alleles cause earlier onset, greater disease severity and faster disease progression. The HTT CAG repeat is genetically unstable in the soma in a process that preferentially generates somatic expansions, the proportion of which is associated with disease onset, severity and progression. Somatic mosaicism of the HTT CAG repeat has traditionally been assessed by semi-quantitative PCR-electrophoresis approaches that have limitations (e.g., no information about sequence variants). Genotyping-by-sequencing could allow for some of these limitations to be overcome.\ud \ud Objective:\ud To investigate the utility of PCR sequencing to genotype large (>50 CAGs) HD alleles and to quantify the associated somatic mosaicism.\ud \ud Methods:\ud We have applied MiSeq and PacBio sequencing to PCR products of the HTT CAG repeat in transgenic R6/2 mice carrying ∼55, ∼110, ∼255 and ∼470 CAGs. For each of these alleles, we compared the repeat length distributions generated for different tissues at two ages.\ud \ud Results:\ud We were able to sequence the CAG repeat full length in all samples. However, the repeat length distributions for samples with ∼470 CAGs were biased towards shorter repeat lengths.\ud \ud Conclusion:\ud PCR sequencing can be used to sequence all the HD alleles considered, but this approach cannot be used to estimate modal allele size or quantify somatic expansions for alleles ⪢250 CAGs. We review the limitations of PCR sequencing and alternative approaches that may allow the quantification of somatic contractions and very large somatic expansions.
- Published
- 2021
18. Mutations causing Lopes-Maciel-Rodan syndrome are huntingtin hypomorphs
- Author
-
Roy Jung, Seung Kwak, Jonathan Picker, Tammy Gillis, Diane Lucente, Douglas Barker, Baehyun Shin, David Howland, Ramee Lee, James F. Gusella, Lance H. Rodan, Marcy E. MacDonald, Yejin Lee, Jong-Min Lee, Jayla Ruliera, Jacob M. Loupe, Ihn Sik Seong, Jayalakshmi S. Mysore, Kevin Correia, and Ryan L. Collins
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,RNA Splicing ,Mutation, Missense ,Biology ,medicine.disease_cause ,Compound heterozygosity ,Cell Line ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Loss of Function Mutation ,mental disorders ,Genetics ,Huntingtin Protein ,medicine ,Humans ,Missense mutation ,Amino Acid Sequence ,RNA, Messenger ,Child ,Molecular Biology ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Mutation ,Heterozygote advantage ,Sequence Analysis, DNA ,General Medicine ,Phenotype ,Pedigree ,nervous system diseases ,Gene Expression Regulation ,nervous system ,Neurodevelopmental Disorders ,Child, Preschool ,Female ,General Article ,Sequence Alignment ,030217 neurology & neurosurgery - 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.
- Published
- 2021
19. Modulation of huntingtin degradation by cAMP-dependent protein kinase A (PKA) phosphorylation of C-HEAT domain Ser2550
- Author
-
Yejin Lee, Hyeongju Kim, Douglas Barker, Ravi Vijayvargia, Ranjit Singh Atwal, Harrison Specht, Hasmik Keshishian, Steven A Carr, Ramee Lee, Seung Kwak, Kyung-gi Hyun, Jacob Loupe, Marcy E. MacDonald, Ji-Joon Song, and Ihn Sik Seong
- Abstract
Huntington’s disease (HD) is a neurodegerative disorder caused by an inherited unstable HTT CAG repeat that expands further, thereby eliciting a disease process that may be initiated by polyglutamine-expanded huntingtin or a short polyglutamine-product. Phosphorylation of selected candidate residues is reported to mediate polyglutamine-fragment degradation and toxicity. Here to support the discovery of phospho-sites involved in the life-cycle of (full-length) huntingtin, we employed mass spectrometry-based phosphoproteomics to systematically identify sites in purified huntingtin and in the endogenous protein, by proteomic and phospho-proteomic analyses of members of an HD neuronal progenitor cell panel. Our results bring total huntingtin phospho-sites to 95, with more located in the N-HEAT domain relative to numbers in the Bridge and C-HEAT domains. Moreover, phosphorylation of C-HEAT Ser2550 by cAMP-dependent protein kinase (PKA), the top hit in kinase activity screens, was found to hasten huntingtin degradation, such that levels of the catalytic subunit (PRKACA) were inversely related to huntingtin levels. Taken together these findings highlight categories of phospho-sites that merit further study and provide a phospho-site kinase pair (pSer2550-PKA) with which to investigate the biological processes that regulate huntingtin degradation and thereby influence the steady state levels of huntingtin in HD cells.
- Published
- 2022
20. Promotion of somatic CAG repeat expansion by Fan1 knock-out in Huntington’s disease knock-in mice is blocked by Mlh1 knock-out
- Author
-
Vanessa C. Wheeler, Ramee Lee, Marina Kovalenko, Kyung Hee Kim, Ricardo Mouro Pinto, Ihn Sik Seong, Tammy Gillis, Marissa A Andrew, Marcy E. MacDonald, Seung Kwak, James F. Gusella, Jacob M. Loupe, Jayalakshmi S. Mysore, Ryan Murtha, Jong-Min Lee, and David Howland
- Subjects
AcademicSubjects/SCI01140 ,congenital, hereditary, and neonatal diseases and abnormalities ,Somatic cell ,Cell Cycle Proteins ,Biology ,MLH1 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Gene interaction ,Huntington's disease ,Gene knockin ,Ribonucleotide Reductases ,mental disorders ,Genetics ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Molecular Biology ,Gene ,Genetics (clinical) ,030304 developmental biology ,Mice, Knockout ,Huntingtin Protein ,0303 health sciences ,Endodeoxyribonucleases ,Genes, Modifier ,General Medicine ,medicine.disease ,Multifunctional Enzymes ,Phenotype ,nervous system diseases ,Disease Models, Animal ,Exodeoxyribonucleases ,Huntington Disease ,General Article ,MutL Protein Homolog 1 ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Genome-Wide Association Study - 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.
- Published
- 2020
21. Genetic Risk Underlying Psychiatric and Cognitive Symptoms in Huntington’s Disease
- Author
-
Christopher Medway, Vanessa C. Wheeler, Diane Lucente, Amelia Tee, Douglas Barker, Seung Kwak, Anka G Ehrhardt, Jean Paul Vonsattel, Kevin Correia, Michael J. Chao, Jacob M. Loupe, Darren G. Monckton, Alastair Maxwell, Jayalakshmi S. Mysore, Lesley Jones, Michael Orth, Richard H. Myers, Thomas Massey, Hugh Rickards, Ira Shoulson, Timothy Stone, Kyung Hee Kim, Marcy E. MacDonald, Marc Ciosi, Branduff McAllister, Erik van Duijn, Lynsey Hall, Tammy Gillis, Duncan McLauchlan, Jong-Min Lee, Ricardo Mouro Pinto, James F. Gusella, Eliana Marisa Ramos, Eun Pyo Hong, Jane S. Paulsen, Peter Holmans, G. Bernhard Landwehrmeyer, Cristina Sampaio, Afroditi Chatzi, Natalie Ellis, Kawther Abu Elneel, and Jeffrey D. Long
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Psychosis ,Population ,Disease ,Irritability ,Article ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Huntington's disease ,Risk Factors ,Genetic variation ,Humans ,Medicine ,Apathy ,Cognitive decline ,Psychiatry ,education ,Biological Psychiatry ,Depression (differential diagnoses) ,030304 developmental biology ,Genetic association ,0303 health sciences ,education.field_of_study ,Depression ,business.industry ,Polygenic risk ,medicine.disease ,3. Good health ,Huntington Disease ,030104 developmental biology ,Psychotic Disorders ,Schizophrenia ,Psychiatric ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Huntington’s disease - Abstract
Huntington’s disease (HD) is an inherited neurodegenerative disorder caused by an expanded CAG repeat in the HTT gene. It is diagnosed following a standardized exam of motor control and often presents with cognitive decline and psychiatric symptoms. Recent studies have detected genetic loci modifying the age at onset of motor symptoms in HD, but genetic factors influencing cognitive and psychiatric presentations are unknown. We tested the hypothesis that psychiatric and cognitive symptoms in HD are influenced by the same common genetic variation as in the general population by constructing polygenic risk scores from large genome-wide association studies of psychiatric and neurodegenerative disorders, and of intelligence, and testing for correlation with the presence of psychiatric and cognitive symptoms in a large sample (n=5160) of HD patients. Polygenic risk score for major depression was associated specifically with increased risk of depression in HD, as was schizophrenia risk score with psychosis and irritability. Cognitive impairment and apathy were associated with reduced polygenic risk score for intelligence. In general, polygenic risk scores for psychiatric disorders, particularly depression and schizophrenia, are associated with increased risk of the corresponding psychiatric symptoms in HD, suggesting a common genetic liability. However, the genetic liability to cognitive impairment and apathy appears to be distinct from other psychiatric symptoms in HD. No associations were observed between HD symptoms and risk scores for other neurodegenerative disorders. These data provide a rationale for treatments effective in depression and schizophrenia to be used to treat depression and psychotic symptoms in HD.
- Published
- 2020
22. Genetic modifiers of Huntington’s disease differentially influence motor and cognitive domains
- Author
-
Jong-Min Lee, Yuan Huang, Michael Orth, Tammy Gillis, Jacqueline Siciliano, Eunpyo Hong, Jayalakshmi Srinidhi Mysore, Diane Lucente, Vanessa C. Wheeler, Ihn Sik Seong, Zachariah L. McLean, James A. Mills, Branduff McAllister, Sergey V. Lobanov, Thomas H. Massey, Marc Ciosi, G. Bernhard Landwehrmeyer, Jane S. Paulsen, E. Ray Dorsey, Ira Shoulson, Cristina Sampaio, Darren G. Monckton, Seung Kwak, Peter Holmans, Lesley Jones, Marcy E. MacDonald, Jeffrey D. Long, and James F. Gusella
- Abstract
Genome-wide association studies (GWAS) of Huntington’s 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 using common motor and cognitive measures to predict age at other disease landmarks as additional phenotypes for GWAS. Combined with imputation using 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 new modifier effects at the PMS1 and PMS2 loci and implicated a potential new 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.
- Published
- 2022
23. Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington’s disease
- Author
-
Yasaman Ataei, Larry Park, D. James Surmeier, B. Joseph Vu, Seung Kwak, Richard T. Surosky, Anand Narayanan, David A. Shivak, Josee Laganiere, Christer Halldin, Andrea Varrone, Matthew C. Mendel, Karsten Tillack, Lei Zhang, Bryan Zeitler, Dmitry Guschin, Lexi Kopan, Sarah J. Hinkley, Kimberly Marlen, Jocelynn R. Pearl, Qi Yu, Taneli Heikkinen, Annette Gärtner, Yalda Sedaghat, Christina Thiede, Miklós Tóth, Jennifer M. Cherone, David Paschon, Jyothisri Kondapalli, Andrea E. Kudwa, Ladislav Mrzljak, Rainier Amora, Kimmo Lehtimäki, Edward J. Rebar, Lenke Tari, Ignacio Munoz-Sanjuan, Jeffrey C. Miller, Sylvie Ramboz, Marie Svedberg, Steven Froelich, Irina Ankoudinova, Philip D. Gregory, Stephen Lam, Michelle Day, Jonathan Bard, Hoang Oanh B. Nguyen, Fyodor D. Urnov, Davis Li, Jenny Haggkvist, H. Steve Zhang, and Guijuan Qiao
- Subjects
0301 basic medicine ,Zinc finger ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutant ,General Medicine ,Biology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,nervous system diseases ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Huntington's disease ,Transcription (biology) ,030220 oncology & carcinogenesis ,mental disorders ,medicine ,Gene silencing ,Allele ,Gene ,Transcription factor - Abstract
Huntington’s disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress >99% of HD-causing alleles over a wide dose range while preserving expression of >86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD. Zinc finger protein transcription factors are developed for the selective silencing of the mutant huntingtin gene in human neurons in vitro and multiple animal models of Huntington’s disease in vivo while preserving expression of the wild-type allele.
- Published
- 2019
24. Association analysis of chromosome X to identify genetic modifiers of Huntington's disease
- Author
-
Marcy E. MacDonald, Michael J. Chao, Eun Pyo Hong, Darren G. Monckton, Vanessa C. Wheeler, Thomas Massey, Diane Lucente, Marc Ciosi, James F. Gusella, Seung Kwak, Michael Orth, Lesley Jones, Jeffrey D. Long, Peter Holmans, S. V. Lobanov, Jong-Min Lee, and Branduff McAllister
- Subjects
0301 basic medicine ,Male ,Huntingtin ,X Chromosome ,DNA repair ,Single-nucleotide polymorphism ,Biology ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Huntington's disease ,Genetic variation ,medicine ,Humans ,Age of Onset ,X chromosome ,Genetic association ,Genetics ,Huntingtin Protein ,Genes, Modifier ,medicine.disease ,030104 developmental biology ,Huntington Disease ,Female ,Neurology (clinical) ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Background: Huntington’s disease (HD) is caused by an expanded (>35) CAG trinucleotide repeat in huntingtin (HTT). Age-at-onset of motor symptoms is inversely correlated with the size of the inherited CAG repeat, which expands further in brain regions due to somatic repeat instability. Our recent genetic investigation focusing on autosomal SNPs revealed that age-at-onset is also influenced by genetic variation at many loci, the majority of which encode genes involved in DNA maintenance/repair processes and repeat instability. Objective: We performed a complementary association analysis to determine whether variants in the X chromosome modify HD. Methods: We imputed SNPs on chromosome X for ∼9,000 HD subjects of European ancestry and performed an X chromosome-wide association study (XWAS) to test for association with age-at-onset corrected for inherited CAG repeat length. Results: In a mixed effects model XWAS analysis of all subjects (males and females), assuming random X-inactivation in females, no genome-wide significant onset modification signal was found. However, suggestive significant association signals were detected at Xq12 (top SNP, rs59098970; p-value, 1.4E-6), near moesin (MSN), in a region devoid of DNA maintenance genes. Additional suggestive signals not involving DNA repair genes were observed in male- and female-only analyses at other locations. Conclusion: Although not genome-wide significant, potentially due to small effect size compared to the power of the current study, our data leave open the possibility of modification of HD by a non-DNA repair process. Our XWAS results are publicly available at the updated GEM EURO 9K website hosted at https://www.hdinhd.org/ for browsing, pathway analysis, and data download.
- Published
- 2021
25. Astaxanthin Inhibits Interleukin-6 Expression in Cerulein/Resistin-Stimulated Pancreatic Acinar Cells
- Author
-
Hyeyoung Kim, Min Seung Kwak, and Joo Weon Lim
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Article Subject ,Immunology ,Anti-Inflammatory Agents ,Acinar Cells ,Xanthophylls ,medicine.disease_cause ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Adipokines ,Internal medicine ,medicine ,Pathology ,RB1-214 ,Animals ,Resistin ,Obesity ,Interleukin 6 ,Pancreas ,Chelating Agents ,NADPH oxidase ,biology ,Chemistry ,Interleukin-6 ,NADPH Oxidase 1 ,NADPH Oxidases ,Cell Biology ,medicine.disease ,Rats ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,030220 oncology & carcinogenesis ,biology.protein ,Acute pancreatitis ,Calcium ,Reactive Oxygen Species ,Intracellular ,Oxidative stress ,Ceruletide ,Research Article - Abstract
Acute pancreatitis is a common clinical condition with increasing the proinflammatory mediators, including interleukin-6 (IL-6). Obesity is a negative prognostic factor in acute pancreatitis. Obese patients with acute pancreatitis have a higher systemic inflammatory response rate. Levels of serum resistin, an adipocytokine secreted by fat tissues, increase with obesity. Cerulein, a cholecystokinin analog, induces calcium (Ca2+) overload, oxidative stress, and IL-6 expression in pancreatic acinar cells, which are hallmarks of acute pancreatitis. A recent study showed that resistin aggravates the expression of inflammatory cytokines in cerulein-stimulated pancreatic acinar cells. We aimed to investigate whether resistin amplifies cerulein-induced IL-6 expression and whether astaxanthin (ASX), an antioxidant carotenoid with anti-inflammatory properties, inhibits ceruelin/resistin-induced IL-6 expression in pancreatic acinar AR42J cells. We found that resistin enhanced intracellular Ca2+ levels, NADPH oxidase activity, intracellular reactive oxygen species (ROS) production, NF-κB activity, and IL-6 expression in cerulein-stimulated AR42J cells, which were inhibited by ASX in a dose-dependent manner. The calcium chelator BAPTA-AM inhibited cerulein/resistin-induced NADPH oxidase activation and ROS production. Antioxidant N-acetyl cysteine (NAC) and ML171, a specific NADPH oxidase 1 inhibitor, suppressed cerulein/resistin-induced ROS production, NF-κB activation, and IL-6 expression. In conclusion, ASX inhibits IL-6 expression, by reducing Ca2+ overload, NADPH oxidase-mediated ROS production, and NF-κB activity in cerulein/resistin-stimulated pancreatic acinar cells. Consumption of ASX-rich foods could be beneficial for preventing or delaying the incidence of obesity-associated acute pancreatitis.
- Published
- 2021
- Full Text
- View/download PDF
26. DNA methylation study of Huntington's disease and motor progression in patients and in animal models
- Author
-
Skye R. Rudiger, X. William Yang, Darren G. Monckton, Alicia Preiss, Russell G. Snell, Jim Rosinski, Hilary Wilkinson, Steve Horvath, Jian Chen, Timothy A. Hore, C. Simon Bawden, Alastair Maxwell, Peter Langfelder, Matthew J Grant, Seung Kwak, Afroditi Chatzi, Jeff Aaronson, Pritika Narayan, Nan Wang, Richard Z. Chen, Giovanni Coppola, Thomas F. Vogt, Ake T. Lu, and Marc Ciosi
- Subjects
0301 basic medicine ,Huntington's Disease ,Male ,Epigenomics ,Huntingtin ,General Physics and Astronomy ,Neurodegenerative ,medicine.disease_cause ,Severity of Illness Index ,Epigenesis, Genetic ,Global Burden of Disease ,Animals, Genetically Modified ,Mice ,0302 clinical medicine ,80 and over ,2.1 Biological and endogenous factors ,Registries ,Longitudinal Studies ,Prospective Studies ,Gene Knock-In Techniques ,Aetiology ,lcsh:Science ,Aged, 80 and over ,Mutation ,Huntingtin Protein ,Multidisciplinary ,DNA methylation ,Behavior, Animal ,Genetic disorder ,Huntington's disease ,Methylation ,Middle Aged ,Recombinant Proteins ,Huntington Disease ,CpG site ,Neurological ,Disease Progression ,Female ,Epigenetics ,Adult ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Adolescent ,Science ,Genetically Modified ,and over ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Genetic ,Internal medicine ,mental disorders ,medicine ,Genetics ,Animals ,Humans ,Aged ,Behavior ,Sheep ,Animal ,Human Genome ,Neurosciences ,General Chemistry ,DNA Methylation ,medicine.disease ,Brain Disorders ,nervous system diseases ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Orphan Drug ,Cross-Sectional Studies ,nervous system ,Genetic Loci ,Disease Models ,CpG Islands ,lcsh:Q ,sense organs ,030217 neurology & neurosurgery ,Follow-Up Studies ,Epigenesis ,Genome-Wide Association Study - Abstract
Although Huntington’s disease (HD) is a well studied Mendelian genetic disorder, less is known about its associated epigenetic changes. Here, we characterize DNA methylation levels in six different tissues from 3 species: a mouse huntingtin (Htt) gene knock-in model, a transgenic HTT sheep model, and humans. Our epigenome-wide association study (EWAS) of human blood reveals that HD mutation status is significantly (p, Although Huntington’s disease (HD) is a well-studied genetic disorder, less is known about the epigenetic changes underlying it. Here, the authors characterize DNA methylation levels in tissues from patients, a mouse huntingtin (Htt) gene knock-in model, and a transgenic HTT sheep model, and provide evidence that HD is accompanied by DNA methylation changes in these three species.
- Published
- 2020
27. Huntingtin lowering reduces somatic instability at CAG-expanded loci
- Author
-
Holly B. Kordasiewicz, Vanessa C. Wheeler, Marissa A Andrew, Michael Flower, Deanna Marchionini, Seung Kwak, José M. Carrillo, Cassandra A. McHugh, Heather Ging, Robert M. Bragg, Julie-Anne Rodier, Jeffrey P. Cantle, Sydney R. Coffey, Scott Zeitlin, David Howland, Hilary Wilkinson, C. Frank Bennett, Ricardo Mouro Pinto, Marina Kovalenko, Sarah J. Tabrizi, Georg Auburger, Joseph Hamilton, and Jeffrey B. Carroll
- Subjects
Genetics ,Pathogenesis ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,nervous system ,Somatic cell ,animal diseases ,mental disorders ,Huntingtin Protein ,Disease ,Biology ,nervous system diseases - Abstract
Expanded trinucleotide repeats cause many human diseases, including Huntington’s disease (HD). Recent studies indicate that somatic instability of these repeats contributes to pathogenesis in several expansion disorders. We find that lowering huntingtin protein (HTT) levels reduces somatic instability of both the Htt and Atxn2 CAG tracts in knockin mouse models, and the HTT CAG tract in human iPSC-derived neurons, revealing an unexpected role for HTT in regulating somatic instability.
- Published
- 2020
28. Genetic and functional analyses point to FAN1 as the source of multiple Huntington Disease modifier effects
- Author
-
Marcy E. MacDonald, Jeffrey D. Long, Jongmin Lee, Michael J. Chao, Peter Holmans, Tammy Gillis, Jayalakshmi S. Mysore, Jacob M. Loupe, Michael Orth, Lesley Jones, Darren G. Monckton, Kyung Hee Kim, Jun-Wan Shin, Eun Pyo Hong, Seung Kwak, James F. Gusella, and Ramee Lee
- Subjects
0301 basic medicine ,DNA repair ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,Genetic variation ,Genetics ,medicine ,Humans ,Allele ,Gene ,Genetics (clinical) ,Endodeoxyribonucleases ,FAN1 ,medicine.disease ,Multifunctional Enzymes ,Exodeoxyribonucleases ,HEK293 Cells ,Huntington Disease ,030104 developmental biology ,Haplotypes ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
A recent genome-wide association study of Huntington’s disease (HD) implicated genes involved in DNA \ud maintenance processes as modifiers of onset, including multiple genome-wide significant signals in a chr15 \ud region containing the DNA repair gene FAN1. Here, we have carried out detailed genetic, molecular and \ud cellular investigation of the modifiers at this locus. We find that missense changes within or near the DNA \ud binding domain (p.Arg507His and p.Arg377Trp) reduce FAN1's DNA binding activity and its capacity to rescue \ud mitomycin C-induced cytotoxicity, accounting for two infrequent onset-hastening modifier signals. We also \ud identified a third onset-hastening modifier signal whose mechanism of action remains uncertain but does not \ud involve an amino acid change in FAN1. We present additional evidence that a frequent onset-delaying modifier \ud signal does not alter FAN1 coding sequence but is associated with increased FAN1 mRNA expression in the \ud cerebral cortex. Consistent with these findings and other cellular overexpression/suppression studies, knock\ud out of FAN1 increased CAG repeat expansion in HD induced pluripotent stem cells. Together, these studies \ud support the process of somatic CAG repeat expansion as a therapeutic target in HD, and clearly indicate that \ud multiple genetic variations act by different means through FAN1 to influence HD onset in a manner that is \ud largely additive, except in the rare circumstance that two onset-hastening alleles are present. Thus, an \ud individual’s particular combination of FAN1 haplotypes may influence their suitability for HD clinical trials, \ud particularly if the therapeutic agent aims to reduce CAG repeat instability.
- Published
- 2020
29. Which Patients Are a Better Candidate of Laparoscopic Repair in Obturator Hernia Patients?
- Author
-
Sang Eok Lee, Si Min Park, In Seok Choi, In Eui Bae, Seong Uk Kwon, Jae Seung Kwak, Ju Ik Moon, Nak Song Sung, Seung Jae Lee, Dae Sung Yoon, and Won Jun Choi
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,medicine ,Obturator hernia ,Laparoscopy ,medicine.disease ,business ,Hernia repair ,Surgery - Abstract
Obturator hernia is a difficult disease to diagnose. If a surgical treatment is delayed in obturator hernia, a bowel resection may be required due to strangulation. The surgical treatment of this disease is to use a classical laparotomy. Recently, the laparoscopic approach has been reported and reviewed for efficiency. We checked the indicators that determine the most appropriate surgical method according to the patient's condition.In the study, a single-institution, retrospective analysis of surgical patients undergoing an obturator hernia surgery between 2003 and 2018 was performed. The patients were divided into a laparoscopic group (5 patients underwent laparoscopic repair; no intestinal resection) and an open group (13 patients who underwent open repair; 10 with and 3 without intestinal resection). The outcomes were compared between the groups. We analyzed the relevant factors that could predict the proper method of surgery.A total 18 patients were included in the study. All patients were female, with body mass index (BMI) of under 21 kg/mObturator hernia can be treated with a laparoscopic surgery. The choice of surgical treatment can be considered in advance through the review of the patient's WBC count or CRP count.
- Published
- 2020
30. Novel DNA Aptamers that Bind to Mutant Huntingtin and Modify Its Activity
- Author
-
Hyeongseok Lee, Ramee Lee, Gwen E. Owens, Roy Jung, Seung Kwak, Ihn Sik Seong, Baehyun Shin, Ji-Joon Song, Jong-Min Lee, Hye Jin Oh, Susan L. Cotman, Ravi Vijayvargia, and Marcy E. MacDonald
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Aptamer ,full-length huntingtin ,Mutant ,single-stranded oligonucleotide ,medicine.disease_cause ,polycomb repressive complex 2 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Mutant protein ,mental disorders ,Drug Discovery ,medicine ,Mutation ,Chemistry ,lcsh:RM1-950 ,Huntington's disease ,Transfection ,Polyglutamine tract ,nervous system diseases ,Cell biology ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,HEAT repeats ,Molecular Medicine ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery - Abstract
The CAG repeat expansion that elongates the polyglutamine tract in huntingtin is the root genetic cause of Huntington’s Disease (HD), a debilitating neurodegenerative disorder. This seemingly slight change to the primary amino acid sequence alters the physical structure of the mutant protein and alters its activity. We have identified a set of G-quadruplex-forming DNA aptamers (MS1, MS2, MS3, MS4) that bind mutant huntingtin proximal to lysines K2932/K2934 in the carboxyl-terminal CTD-II domain. Aptamer-binding to mutant huntingtin, abrogated the enhanced polycomb repressive complex 2 (PRC2) stimulatory-activity conferred by the expanded polyglutamine tract. In HD, but not normal, neuronal progenitor cells (NPC), MS3 aptamer co-localized with endogenous mutant huntingtin and was associated with significantly decreased PRC2 activity. Furthermore, MS3 transfection protected HD NPC against starvation-dependent stress with increased ATP. Therefore, DNA aptamers can preferentially target mutant huntingtin and modulate a gain of function endowed by the elongated polyglutamine segment. These mutant huntingtin binding aptamers provide novel molecular tools for delineating the effects of the HD mutation and encourage mutant huntingtin structure-based approaches to therapeutic development.
- Published
- 2018
31. Full sequence of mutant huntingtin 3'-untranslated region and modulation of its gene regulatory activity by endogenous microRNA
- Author
-
Ramee Lee, Kyung Hee Kim, Marcy E. MacDonald, Kawther Abu Elneel, James F. Gusella, Jun-Wan Shin, Ihn Sik Seong, David Seong, Jae Whan Keum, Jong-Min Lee, and Seung Kwak
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Mutant ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Exon ,Genes, Reporter ,mental disorders ,Genetics ,medicine ,Humans ,Allele ,Gene ,3' Untranslated Regions ,Genetics (clinical) ,Alleles ,Mutation ,Huntingtin Protein ,Three prime untranslated region ,nervous system diseases ,MicroRNAs ,030104 developmental biology ,HEK293 Cells ,Huntington Disease ,nervous system ,Gene Expression Regulation ,Haplotypes ,Trinucleotide repeat expansion - Abstract
Huntington’s disease (HD) is caused by an expanded CAG trinucleotide repeat in the first exon of the huntingtin gene (HTT). Since the entire course of the disease starts from this dominant gain-of-function mutation, lowering total or mutant huntingtin mRNA/protein has emerged as an appealing therapeutic strategy. We reasoned that endogenous mechanisms underlying HTT gene regulation may inform strategies to target the source of the disease. As part of our investigation to understand how the expression of HTT is controlled, we performed (1) complete sequencing analysis for mutant HTT 3′-UTR and (2) unbiased screening assays to identify naturally-occurring miRNAs that could lower the HTT mRNA levels. By sequencing HD families inheriting the major European mutant haplotype, we determined the full sequence of HTT 3′-UTRs of the most frequent mutant (i.e., hap.01) and normal (i.e., hap.08) haplotypes, revealing 5 sites with alternative alleles. In subsequent miRNA activity assays using the full-length hap.01 and hap.08 3′-UTR reporter vectors and follow-up validation experiments, hsa-miR-4324 and hsa-miR-4756-5p significantly reduced HTT 3′-UTR reporter activity and endogenous HTT protein levels. However, those miRNAs did not show strong haplotype-specific effects. Nevertheless, our data highlighting full sequences of HTT 3′-UTR haplotypes, effects of miRNAs on HTT levels, and potential interaction sites provide rationale and promising targets for total and mutant-specific HTT lowering intervention strategies using endogenous and artificial miRNAs, respectively.
- Published
- 2019
32. Huntington’s disease onset is determined by length of uninterrupted CAG, not encoded polyglutamine, and is modified by DNA maintenance mechanisms
- Author
-
Diane Lucente, Douglas Barker, Michael J. Chao, Branduff McAllister, Georg Bernhard Landwehrmeyer, Abu Elneel K, Lynsey S. Hall, Jean-Paul Vonsattel, Jacob M. Loupe, Anka G Ehrhardt, J.S. Paulsen, Richard H. Myers, Vanessa C. Wheeler, Christopher W Medway, Jayalakshmi S. Mysore, Alastair Maxwell, Ira Shoulson, Tammy Gillis, Seung Kwak, Michael Orth, Peter Holmans, Timothy Stone, Lesley Jones, Eliana Marisa Ramos, J. F. Gusella, Cristina Sampaio, Eun Pyo Hong, Julianna Y. Lee, Dorsey Er, Mouro Pinto R, Kyuseok Kim, Kevin Correia, Jeffrey D. Long, Afroditi Chatzi, Tom Massey, Marcy E. MacDonald, Darren G. Monckton, and Marc Ciosi
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Somatic cell ,Repeat sequence ,Genome-wide association study ,Biology ,medicine.disease ,Pathogenesis ,Huntington's disease ,DNA Maintenance ,mental disorders ,medicine ,Gene - Abstract
SUMMARYThe effects of variable, glutamine-encoding, CAA interruptions indicate that a property of the uninterrupted HTT CAG repeat sequence, distinct from huntingtin’s polyglutamine segment, dictates the rate at which HD develops. The timing of onset shows no significant association with HTT cis-eQTLs but is influenced, sometimes in a sex-specific manner, by polymorphic variation at multiple DNA maintenance genes, suggesting that the special onset-determining property of the uninterrupted CAG repeat is a propensity for length instability that leads to its somatic expansion. Additional naturally-occurring genetic modifier loci, defined by GWAS, may influence HD pathogenesis through other mechanisms. These findings have profound implications for the pathogenesis of HD and other repeat diseases and question a fundamental premise of the “polyglutamine disorders”.
- Published
- 2019
33. Huntington's disease accelerates epigenetic aging of human brain and disrupts DNA methylation levels
- Author
-
Giovanni Coppola, Marika Eszes, Seung Kwak, Richard L.M. Faull, Spencer Tung, Jim Rosinski, Oi-Wa Choi, Thomas F. Vogt, Peter Langfelder, Steve Horvath, X. William Yang, Henry J. Waldvogel, Maurice A. Curtis, Harry V. Vinters, and Jeff Aaronson
- Subjects
Adult ,Male ,0301 basic medicine ,Aging ,medicine.medical_specialty ,Adolescent ,brain ,Biology ,biomarker of aging ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,Internal medicine ,medicine ,Humans ,Epigenetics ,Age of Onset ,Aged ,Genetics ,DNA methylation ,epigenetics ,Age Factors ,Cell Biology ,Methylation ,Human brain ,Middle Aged ,medicine.disease ,Huntington Disease ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Frontal lobe ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,Age of onset ,epigenetic clock ,Research Paper - Abstract
Age of Huntington's disease (HD) motoric onset is strongly related to the number of CAG trinucleotide repeats in the huntingtin gene, suggesting that biological tissue age plays an important role in disease etiology. Recently, a DNA methylation based biomarker of tissue age has been advanced as an epigenetic aging clock. We sought to inquire if HD is associated with an accelerated epigenetic age. DNA methylation data was generated for 475 brain samples from various brain regions of 26 HD cases and 39 controls. Overall, brain regions from HD cases exhibit a significant epigenetic age acceleration effect (p=0.0012). A multivariate model analysis suggests that HD status increases biological age by 3.2 years. Accelerated epigenetic age can be observed in specific brain regions (frontal lobe, parietal lobe, and cingulate gyrus). After excluding controls, we observe a negative correlation (r=−0.41, p=5.5×10−8) between HD gene CAG repeat length and the epigenetic age of HD brain samples. Using correlation network analysis, we identify 11 co-methylation modules with a significant association with HD status across 3 broad cortical regions. In conclusion, HD is associated with an accelerated epigenetic age of specific brain regions and more broadly with substantial changes in brain methylation levels.
- Published
- 2016
34. Large-scale phenome analysis defines a behavioral signature for Huntington's disease genotype in mice
- Author
-
Igor Filipov, Vanessa C. Wheeler, Matthew J. Mazzella, Liliana B. Menalled, Melinda C Ruiz, Ana Sanchez, Sylvie Ramboz, Brenda Lager, Marcy E. MacDonald, Ian Russell, Kimberly Cox, Miguel A. Gomez, Afshin Ghavami, Vadim Alexandrov, Seung Kwak, Dani Brunner, Justin Torello, Jeff Aaronson, Mei Kwan, Andrea E. Kudwa, James F. Gusella, Judy Watson-Johnson, Jim Rosinski, David Howland, and Emily Sabath
- Subjects
0301 basic medicine ,Biomedical Engineering ,Bioengineering ,Genome-wide association study ,Computational biology ,Phenome ,Biology ,Polymorphism, Single Nucleotide ,Applied Microbiology and Biotechnology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,Genetic variation ,Genotype ,medicine ,Huntingtin Protein ,Animals ,Genetics ,Genome ,Behavior, Animal ,Neurodegeneration ,Chromosome Mapping ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Huntington Disease ,Phenotype ,030104 developmental biology ,Molecular Medicine ,Age of onset ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Biotechnology - Abstract
Rapid technological advances for the frequent monitoring of health parameters have raised the intriguing possibility that an individual's genotype could be predicted from phenotypic data alone. Here we used a machine learning approach to analyze the phenotypic effects of polymorphic mutations in a mouse model of Huntington's disease that determine disease presentation and age of onset. The resulting model correlated variation across 3,086 behavioral traits with seven different CAG-repeat lengths in the huntingtin gene (Htt). We selected behavioral signatures for age and CAG-repeat length that most robustly distinguished between mouse lines and validated the model by correctly predicting the repeat length of a blinded mouse line. Sufficient discriminatory power to accurately predict genotype required combined analysis of >200 phenotypic features. Our results suggest that autosomal dominant disease-causing mutations could be predicted through the use of subtle behavioral signatures that emerge in large-scale, combinatorial analyses. Our work provides an open data platform that we now share with the research community to aid efforts focused on understanding the pathways that link behavioral consequences to genetic variation in Huntington's disease.
- Published
- 2016
35. Ultrarapid, size-controlled, high-crystalline plasma-mediated synthesis of ceria nanoparticles for reagent-free colorimetric glucose test strips
- Author
-
J. S. Park, Ho Seung Kwak, Young-Seak Lee, Hyeran Kim, Thao Nguyen Le, Ha-Rim An, Soon Chang Lee, Yesul Jeong, Yong Cheol Hong, Kyuseok Choi, Sang-Gil Lee, Hyun Uk Lee, Xuan Ai Le, Moon Sang Lee, Hee Tae Ahn, Seung Jo Yoo, Young-Boo Lee, and Moon Il Kim
- Subjects
medicine.diagnostic_test ,Chemistry ,Metals and Alloys ,Nanoparticle ,Plasma ,STRIPS ,Condensed Matter Physics ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,law.invention ,law ,Reagent ,Materials Chemistry ,medicine ,Glucose test ,Electrical and Electronic Engineering ,Instrumentation ,Nuclear chemistry - Published
- 2020
36. F74 Origin-hd: genetic modifiers of htt cag intergenerational repeat instability in male hdgecs
- Author
-
Seung Kwak, Adam Ellenberger, Edward J. Wild, Gail Owen, Anka G Ehrhardt, Elena Pak, Sherry Lifer, and Cristina Sampaio
- Subjects
Genetics ,DNA repair ,Somatic cell ,Anticipation (genetics) ,Disease ,Allele ,Biology ,Germline ,Genotype frequency ,Genetic association - Abstract
Background Earlier disease onset in subsequent generations, called anticipation, has been observed in HD families and is attributed to an increased CAG-repeat length in the HTT gene. CAG-repeat length mutations are referred to as ‘repeat instability’ and genome-wide association studies suggest that genomic variants function as genetic modifiers of disease onset, for example by affecting DNA repair mechanisms. Aims Origin-HD aims to investigate CAG repeat instability in germline and somatic cells and evaluate for correlations with putative genetic modifiers. Understanding the mechanisms affecting repeat instability may yield testable targets for future interventions. Study design Starting Q2 2019, over 1,000 male HDGECs (ages 18–55) will be recruited from Enroll-HD participants over about two years, approximately balanced between premanifest and manifest disease. Allele and genotype frequency for each pre-specified variant of interest will be assessed after about 500 participants have been recruited. If the predicted detectable effect size is d>0.35 in the final sample of 1,000 participants, a recruitment-by-genotype recruitment approach can be adopted in parallel to the ongoing recruitment. The repeat instability in DNA from sperm and blood of study participants will be analyzed, and genetic modifier variants will be determined.
- Published
- 2018
37. C01 Glutamine codon usage and somatic mosaicism of the HTT cag repeat are modifiers of huntington disease severity
- Author
-
Blair R. Leavitt, Alexandra Durr, Peter Holmans, Davina J. Hensman Moss, Marc Ciosi, Michael Flower, Sarah J. Tabrizi, Darren G. Monckton, Seung Kwak, Raymund A.C. Roos, Alastair Maxwell, Douglas R. Langbehn, Sarah A. Cumming, Asma M. Alshammari, and Lesley Jones
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Exon ,MSH3 ,Somatic cell ,mental disorders ,Genotype ,Genome-wide association study ,MLH3 ,Biology ,Allele ,Genetic association - Abstract
Background Huntington disease (HD) is caused by the expansion of a polyglutamine encoding CAG repeat in exon 1 of the HTT gene. Affected individuals inherit ≥40 repeats and longer alleles are associated with earlier onset and higher HD severity. The HTT CAG repeat is genetically unstable in both the germline and soma. Somatic mosaicism is dependent on the number of CAG repeats and age, and is expansion biased and cell-type specific. Recent genome-wide association studies (GWAS) have identified components of the DNA repair system as trans¬-acting modifiers of HD severity, some of which are known to modify somatic instability of the HTT CAG repeat in HD mouse models. We thus hypothesise that the trans¬-acting modifiers identified by GWAS affect HD severity by their direct effect on HTT CAG somatic instability. Aims Determine the exact trinucleotide structure of the HTT exon 1 repeat and quantify its somatic mosaicism to investigate their association with HD severity. Methods/techniques Using genotyping-by-sequencing, we determined the exact genotype of the polyglutamine and polyproline encoding repeats in HTT exon 1 and quantified the somatic mosaicism associated with the CAG repeat in blood DNA from 807 HD expansion carriers. Results/outcome The sequence encoding the HTT polyglutamine and polyproline tract has an atypical structure in ˜8% of the non-HD-causing alleles and ˜3% of the HD-causing alleles, differing from the typical structure by the number of glutamine encoding CAA codons and/or the number of proline encoding CCA and CCT codons. Multiple linear regression analysis revealed that the number of CAA codons is negatively correlated with HD severity and that the number of CAG repeats is a better predictor of HD severity (r2=0.559) than the number of glutamines (r2=0.537). Moreover, somatic mosaicism in blood correlates with HD severity (r2 ≥0.014, p≤2.5 × 10–3) and some of the polymorphisms associated with HD severity (p=1.5 × 10–5 for FAN1 rs3512, p=1.8 × 10–4 for MLH3 rs175080, p=3.6 × 10–3 for MLH1 rs1799977 and p=0.016 for MSH3 rs1382539). Conclusion Our data show that atypical HD-causing alleles have major implications for genetic diagnosis and counselling and confirm the correlation of somatic expansion with HD severity. The latter further supports the therapeutic potential of targeting expansion causing components of the DNA repair system.
- Published
- 2018
38. Population-specific genetic modification of Huntington's disease in Venezuela
- Author
-
Hong Li, Seung Kwak, Michael Orth, Leroy Hood, Michael J. Chao, Nancy S. Wexler, Diane Lucente, Laura Bannach Jardim, James F. Gusella, Peter Holmans, Jong-Min Lee, Jared C. Roach, Vanessa C. Wheeler, Ju Wan Shin, Marcy E. MacDonald, Kyung Hee Kim, and Lesley Jones
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Heredity ,Idade de início ,Genome-wide association study ,Saúde da família ,QH426-470 ,Geographical Locations ,0302 clinical medicine ,Cell Signaling ,Medicine and Health Sciences ,Age of Onset ,Genetics (clinical) ,Polimorfismo de nucleotídeo único ,Genetics ,Huntingtin Protein ,Intracellular Signaling Peptides and Proteins ,Neurodegenerative Diseases ,Genomics ,Genes modificadores ,Estudo de associação genômica ampla ,3. Good health ,Europe ,Genetic Mapping ,Huntington Disease ,Neurology ,Genetic Diseases ,Medical genetics ,Female ,Haplotipos ,Genetic Engineering ,Genomic Signal Processing ,Research Article ,Biotechnology ,Signal Transduction ,medicine.medical_specialty ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Molecular Genetics ,03 medical and health sciences ,Huntington's disease ,Gene mapping ,medicine ,Genome-Wide Association Studies ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Adaptor Proteins, Signal Transducing ,Clinical Genetics ,Family Health ,Genes, Modifier ,Whole Genome Sequencing ,Haplotype ,Autosomal Dominant Diseases ,Biology and Life Sciences ,Computational Biology ,Proteins ,Proteína huntingtina ,Human Genetics ,Cell Biology ,medicine.disease ,Genome Analysis ,Venezuela ,Human genetics ,030104 developmental biology ,Genetics, Population ,Doença de Huntington ,Haplotypes ,Genetic Loci ,Sequenciamento completo do genoma ,People and Places ,Gene-Environment Interaction ,030217 neurology & neurosurgery ,Genética populacional ,Genome-Wide Association Study - Abstract
Modifiers of Mendelian disorders can provide insights into disease mechanisms and guide therapeutic strategies. A recent genome-wide association (GWA) study discovered genetic modifiers of Huntington's disease (HD) onset in Europeans. Here, we performed whole genome sequencing and GWA analysis of a Venezuelan HD cluster whose families were crucial for the original mapping of the HD gene defect. The Venezuelan HD subjects develop motor symptoms earlier than their European counterparts, implying the potential for population-specific modifiers. The main Venezuelan HD family inherits HTT haplotype hap.03, which differs subtly at the sequence level from European HD hap.03, suggesting a different ancestral origin but not explaining the earlier age at onset in these Venezuelans. GWA analysis of the Venezuelan HD cluster suggests both population-specific and population-shared genetic modifiers. Genome-wide significant signals at 7p21.2–21.1 and suggestive association signals at 4p14 and 17q21.2 are evident only in Venezuelan HD, but genome-wide significant association signals at the established European chromosome 15 modifier locus are improved when Venezuelan HD data are included in the meta-analysis. Venezuelan-specific association signals on chromosome 7 center on SOSTDC1, which encodes a bone morphogenetic protein antagonist. The corresponding SNPs are associated with reduced expression of SOSTDC1 in non-Venezuelan tissue samples, suggesting that interaction of reduced SOSTDC1 expression with a population-specific genetic or environmental factor may be responsible for modification of HD onset in Venezuela. Detection of population-specific modification in Venezuelan HD supports the value of distinct disease populations in revealing novel aspects of a disease and population-relevant therapeutic strategies., Author summary Huntington’s disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in HTT, which encodes huntingtin. HD involves various neurological manifestations such as involuntary movements, cognitive decline, and personality change; age at onset of diagnostic signs is partly determined by the length of the CAG repeat. However, genome-wide analysis of European HD subjects revealed that age at onset corrected for individual CAG repeat size is modified by other genetic factors, revealing potential disease-delaying pathways that form targets for therapeutic strategies. In this study, we set out to identify genetic modifiers in a Venezuelan HD cluster whose families were crucial for discovering the cause of HD. Through genome-wide association analysis using age at onset corrected for CAG repeat size as the phenotype, we found genome-wide significant (chromosome 7) and suggestive significant association signals (chromosomes 4 and 17). Significant modification signals in Venezuelan HD appear to be population-specific because genetic analysis of European HD subjects did not reveal modification signals at those locations. Nevertheless, established modification signals in European HD at a chromosome 15 locus were also augmented by Venezuelan HD data, supporting the existence of both shared and population-specific genetic modifiers in HD. Together with the full sequence of HTT shared by the majority of these Venezuelan HD subjects, our genetic analysis provides insights into the geographical origin of the founder mutation, meaningful allele-specific sites for gene targeting approaches, and potential targets for disease-modifying intervention in this population.
- Published
- 2018
39. MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice
- Author
-
Steve Horvath, Giovanni Coppola, Nan Wang, X. William Yang, Peter Langfelder, David Howland, Seung Kwak, Thomas F. Vogt, Jeffrey S. Aaronson, Jim Rosinski, Fuying Gao, and Li, Xiao-Jiang
- Subjects
0301 basic medicine ,Huntington's Disease ,Huntingtin ,ved/biology.organism_classification_rank.species ,lcsh:Medicine ,Neurodegenerative ,Biochemistry ,Hippocampus ,Transcriptome ,Mice ,Mathematical and Statistical Techniques ,Trinucleotide Repeats ,Cerebellum ,Gene expression ,Medicine and Health Sciences ,lcsh:Science ,Cerebral Cortex ,Huntingtin Protein ,Multidisciplinary ,Brain ,Neurodegenerative Diseases ,Animal Models ,Cell biology ,Nucleic acids ,Huntington Disease ,Neurology ,Experimental Organism Systems ,Genetic Diseases ,Physical Sciences ,Neurological ,Anatomy ,Statistics (Mathematics) ,Research Article ,Biotechnology ,General Science & Technology ,1.1 Normal biological development and functioning ,Mouse Models ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Rare Diseases ,Underpinning research ,microRNA ,Genetics ,Animals ,Humans ,Allele ,Statistical Methods ,Model organism ,Non-coding RNA ,Clinical Genetics ,Messenger RNA ,Biology and life sciences ,ved/biology ,Autosomal Dominant Diseases ,lcsh:R ,Neurosciences ,Gene regulation ,Brain Disorders ,Neostriatum ,MicroRNAs ,030104 developmental biology ,RNA ,lcsh:Q ,Trinucleotide repeat expansion ,Mathematics ,Meta-Analysis - Abstract
In Huntington's disease (HD) patients and in model organisms, messenger RNA transcriptome has been extensively studied; in contrast, comparatively little is known about expression and potential role of microRNAs. Using RNA-sequencing, we have quantified microRNA expression in four brain regions and liver, at three different ages, from an allelic series of HD model mice with increasing CAG length in the endogenous Huntingtin gene. Our analyses reveal CAG length-dependent microRNA expression changes in brain, with 159 microRNAs selectively altered in striatum, 102 in cerebellum, 51 in hippocampus, and 45 in cortex. In contrast, a progressive CAG length-dependent microRNA dysregulation was not observed in liver. We further identify microRNAs whose transcriptomic response to CAG length expansion differs significantly among the brain regions and validate our findings in data from a second, independent cohort of mice. Using existing mRNA expression data from the same animals, we assess the possible relationships between microRNA and mRNA expression and highlight candidate microRNAs that are negatively correlated with, and whose predicted targets are enriched in, CAG-length dependent mRNA modules. Several of our top microRNAs (Mir212/Mir132, Mir218, Mir128 and others) have been previously associated with aspects of neuronal development and survival. This study provides an extensive resource for CAG length-dependent changes in microRNA expression in disease-vulnerable and -resistant brain regions in HD mice, and provides new insights for further investigation of microRNAs in HD pathogenesis and therapeutics.
- Published
- 2018
40. Identification of Genetic Factors that Modify Clinical Onset of Huntington’s Disease
- Author
-
Diane Lucente, Michael J. Chao, Richard H. Myers, Denise Harold, Seung Kwak, Kawther Abu-Elneel, E. Ray Dorsey, Michael Orth, Vanessa C. Wheeler, Ira Shoulson, Timothy Stone, Lesley Jones, Mithra Mahmoudi, Ricardo Mouro Pinto, Tammy Gillis, Marcy E. MacDonald, Jong-Min Lee, Jun Han, Alexey Vedernikov, Eliana Marisa Ramos, Jane S. Paulsen, James F. Gusella, Valentina Escott-Price, Jean Paul G. Vonsattel, Peter Holmans, G. Bernhard Landwehrmeyer, and Jayalakshmi S. Mysore
- Subjects
Locus (genetics) ,Nerve Tissue Proteins ,Disease ,Biology ,MLH1 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Chromosome 15 ,symbols.namesake ,Huntington's disease ,Trinucleotide Repeats ,Genetic variation ,medicine ,Humans ,Age of Onset ,Adaptor Proteins, Signal Transducing ,Genetics ,Chromosomes, Human, Pair 15 ,Huntingtin Protein ,Genes, Modifier ,Biochemistry, Genetics and Molecular Biology(all) ,Nuclear Proteins ,medicine.disease ,Huntington Disease ,Mendelian inheritance ,symbols ,Trinucleotide repeat expansion ,MutL Protein Homolog 1 ,Chromosomes, Human, Pair 8 ,Genome-Wide Association Study - Abstract
SummaryAs a Mendelian neurodegenerative disorder, the genetic risk of Huntington’s disease (HD) is conferred entirely by an HTT CAG repeat expansion whose length is the primary determinant of the rate of pathogenesis leading to disease onset. To investigate the pathogenic process that precedes disease, we used genome-wide association (GWA) analysis to identify loci harboring genetic variations that alter the age at neurological onset of HD. A chromosome 15 locus displays two independent effects that accelerate or delay onset by 6.1 years and 1.4 years, respectively, whereas a chromosome 8 locus hastens onset by 1.6 years. Association at MLH1 and pathway analysis of the full GWA results support a role for DNA handling and repair mechanisms in altering the course of HD. Our findings demonstrate that HD disease modification in humans occurs in nature and offer a genetic route to identifying in-human validated therapeutic targets in this and other Mendelian disorders.PaperClip
- Published
- 2015
- Full Text
- View/download PDF
41. Integration-independent Transgenic Huntington Disease Fragment Mouse Models Reveal Distinct Phenotypes and Life Span in Vivo
- Author
-
Akilah Bonner, Karen Ring, Jennifer Holcomb, Ningzhe Zhang, Robert O’Brien, Khan Zafar, Lisa M. Ellerby, Andreas Weiss, Bradford W. Gibson, Sylvia F. Chen, Brenda Lager, Francesco DeGiacomo, Seung Kwak, and Birgit Schilling
- Subjects
Male ,Genetically modified mouse ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Transgene ,Proteolysis ,Longevity ,Mutant ,Mice, Transgenic ,Nerve Tissue Proteins ,Motor Activity ,Biology ,Biochemistry ,Protein Interaction Mapping ,mental disorders ,medicine ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,Cognitive decline ,Molecular Biology ,Huntingtin Protein ,medicine.diagnostic_test ,HEK 293 cells ,Brain ,Molecular Bases of Disease ,Cell Biology ,Molecular biology ,Phenotype ,nervous system diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,Huntington Disease ,nervous system ,Codon, Nonsense ,Female - Abstract
The cascade of events that lead to cognitive decline, motor deficits, and psychiatric symptoms in patients with Huntington disease (HD) is triggered by a polyglutamine expansion in the N-terminal region of the huntingtin (HTT) protein. A significant mechanism in HD is the generation of mutant HTT fragments, which are generally more toxic than the full-length HTT. The protein fragments observed in human HD tissue and mouse models of HD are formed by proteolysis or aberrant splicing of HTT. To systematically investigate the relative contribution of the various HTT protein proteolysis events observed in vivo, we generated transgenic mouse models of HD representing five distinct proteolysis fragments ending at amino acids 171, 463, 536, 552, and 586 with a polyglutamine length of 148. All lines contain a single integration at the ROSA26 locus, with expression of the fragments driven by the chicken β-actin promoter at nearly identical levels. The transgenic mice N171-Q148 and N552-Q148 display significantly accelerated phenotypes and a shortened life span when compared with N463-Q148, N536-Q148, and N586-Q148 transgenic mice. We hypothesized that the accelerated phenotype was due to altered HTT protein interactions/complexes that accumulate with age. We found evidence for altered HTT complexes in caspase-2 fragment transgenic mice (N552-Q148) and a stronger interaction with the endogenous HTT protein. These findings correlate with an altered HTT molecular complex and distinct proteins in the HTT interactome set identified by mass spectrometry. In particular, we identified HSP90AA1 (HSP86) as a potential modulator of the distinct neurotoxicity of the caspase-2 fragment mice (N552-Q148) when compared with the caspase-6 transgenic mice (N586-Q148).
- Published
- 2015
42. How Middle School Principals Can Affect Beginning Teachers’ Experiences
- Author
-
Ben Pogodzinski, Hyun-Seung Kwak, and Peter Youngs
- Subjects
Medical education ,School administration ,ComputingMilieux_COMPUTERSANDEDUCATION ,Job satisfaction ,Psychology ,Affect (psychology) ,Instructional leadership ,Qualitative research - Abstract
This article reports on a 2-year qualitative research study of the processes by which middle school principals’ policies and actions shaped the experiences of five novice teachers in two Michigan school districts. We examined beginning teachers’ perceptions of principals’ approaches to managing student behavior, instructional leadership, and teacher collaboration and their perceptions of the extent to which each principal was trusted by his or her teaching staff. At the end of the second year of data collection (2007–2008), all five beginning teachers expressed high levels of satisfaction and planned to remain teaching in their schools. We argue that leadership related to student behavior and instruction (as perceived by the novices), combined with high levels of teacher–principal trust (again as perceived by the novices), contributes to these outcomes.
- Published
- 2015
43. A modifier of Huntington's disease onset at the MLH1 locus
- Author
-
Peter Holmans, Denise Harold, Vanessa C. Wheeler, Lesley Jones, Michael Orth, Michael J. Chao, Tammy Gillis, Kawther Abu Elneel, Seung Kwak, Marcy E. MacDonald, Jong-Min Lee, Richard H. Myers, and James F. Gusella
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Genotype ,Genome-wide association study ,Single-nucleotide polymorphism ,Locus (genetics) ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Trinucleotide Repeats ,Huntington's disease ,Genetics ,medicine ,Animals ,Humans ,Allele ,Molecular Biology ,Alleles ,Genetics (clinical) ,Chromosomes, Human, Pair 15 ,Huntingtin Protein ,Genes, Modifier ,Association Studies Articles ,General Medicine ,medicine.disease ,Disease Models, Animal ,Huntington Disease ,Phenotype ,030104 developmental biology ,Expression quantitative trait loci ,MutL Protein Homolog 1 ,030217 neurology & neurosurgery ,Chromosomes, Human, Pair 8 ,Genome-Wide Association Study - Abstract
Huntington’s disease (HD) is a dominantly inherited neurodegenerative disease caused by an expanded CAG repeat in HTT.\ud Many clinical characteristics of HD such as age at motor onset are determined largely by the size of HTT CAG repeat.\ud However, emerging evidence strongly supports a role for other genetic factors in modifying the disease pathogenesis driven\ud by mutant huntingtin. A recent genome-wide association analysis to discover genetic modifiers of HD onset age provided\ud initial evidence for modifier loci on chromosomes 8 and 15 and suggestive evidence for a locus on chromosome 3. Here, genotyping\ud of candidate single nucleotide polymorphisms in a cohort of 3,314 additional HD subjects yields independent confirmation\ud of the former two loci and moves the third to genome-wide significance at MLH1, a locus whose mouse orthologue\ud modifies CAG length-dependent phenotypes in a Htt-knock-in mouse model of HD. Both quantitative and dichotomous association\ud analyses implicate a functional variant on 32% of chromosomes with the beneficial modifier effect that delays HD\ud motor onset by 0.7 years/allele. Genomic DNA capture and sequencing of a modifier haplotype localize the functional variation\ud to a 78 kb region spanning the 3’end of MLH1 and the 5’end of the neighboring LRRFIP2, and marked by an isoleucinevaline\ud missense variant in MLH1. Analysis of expression Quantitative Trait Loci (eQTLs) provides modest support for altered\ud regulation of MLH1 and LRRFIP2, raising the possibility that the modifier affects regulation of both genes. Finally, polygenic\ud modification score and heritability analyses suggest the existence of additional genetic modifiers, supporting expanded, comprehensive\ud genetic analysis of larger HD datasets.
- Published
- 2017
44. Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington's disease
- Author
-
Bryan, Zeitler, Steven, Froelich, Kimberly, Marlen, David A, Shivak, Qi, Yu, Davis, Li, Jocelynn R, Pearl, Jeffrey C, Miller, Lei, Zhang, David E, Paschon, Sarah J, Hinkley, Irina, Ankoudinova, Stephen, Lam, Dmitry, Guschin, Lexi, Kopan, Jennifer M, Cherone, Hoang-Oanh B, Nguyen, Guijuan, Qiao, Yasaman, Ataei, Matthew C, Mendel, Rainier, Amora, Richard, Surosky, Josee, Laganiere, B Joseph, Vu, Anand, Narayanan, Yalda, Sedaghat, Karsten, Tillack, Christina, Thiede, Annette, Gärtner, Seung, Kwak, Jonathan, Bard, Ladislav, Mrzljak, Larry, Park, Taneli, Heikkinen, Kimmo K, Lehtimäki, Marie M, Svedberg, Jenny, Häggkvist, Lenke, Tari, Miklós, Tóth, Andrea, Varrone, Christer, Halldin, Andrea E, Kudwa, Sylvie, Ramboz, Michelle, Day, Jyothisri, Kondapalli, D James, Surmeier, Fyodor D, Urnov, Philip D, Gregory, Edward J, Rebar, Ignacio, Muñoz-Sanjuán, and H Steve, Zhang
- Subjects
Male ,Huntingtin Protein ,Transcription, Genetic ,Zinc Fingers ,Neuroprotection ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Huntington Disease ,Trinucleotide Repeats ,Mutation ,Mice, Inbred CBA ,Animals ,Humans ,Female ,Alleles ,Cells, Cultured - Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress 99% of HD-causing alleles over a wide dose range while preserving expression of 86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD.
- Published
- 2017
45. Novel allele-specific quantification methods reveal no effects of adult onset CAG repeats on HTT mRNA and protein levels
- Author
-
Ihn Sik Seong, Kyung Hee Kim, Aram Shin, Jennifer M. Hope, James F. Gusella, Marcy E. MacDonald, Jong-Min Lee, John D. Leszyk, Tammy Gillis, Scott A. Shaffer, Seung Kwak, Baehyun Shin, Ramee Lee, Ranjit Singh Atwal, and Jun-Wan Shin
- Subjects
0301 basic medicine ,Adult ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutant ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,Genetics ,Huntingtin Protein ,medicine ,Gene silencing ,Humans ,RNA, Messenger ,Allele ,Age of Onset ,Molecular Biology ,Genetics (clinical) ,Alleles ,Regulation of gene expression ,Mutation ,Haplotype ,Brain ,General Medicine ,Articles ,Molecular biology ,030104 developmental biology ,Huntington Disease ,Gene Expression Regulation ,Female ,Autopsy ,Trinucleotide repeat expansion ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery - 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.
- Published
- 2017
46. CAG Repeat Not Polyglutamine Length Determines Timing of Huntington’s Disease Onset
- Author
-
Alastair Maxwell, Richard H. Myers, Michael Orth, Kyung Hee Kim, Kawther Abu Elneel, Lynsey S. Hall, Diane Lucente, Tammy Gillis, Anka G. Ehrhardt, Vanessa C. Wheeler, Christopher Medway, Douglas Barker, Michael J. Chao, Jacob M. Loupe, Darren G. Monckton, Jeffrey D. Long, Thomas Massey, Marc Ciosi, Jayalakshmi S. Mysore, Branduff McAllister, Jongmin Lee, Seung Kwak, E. Ray Dorsey, Kevin Correia, James F. Gusella, Ira Shoulson, Jean Paul G. Vonsattel, Jane S. Paulsen, Peter Holmans, G. Bernhard Landwehrmeyer, Lesley Jones, Eliana Marisa Ramos, Cristina Sampaio, Marcy E. MacDonald, Ricardo Mouro Pinto, Timothy Stone, Afroditi Chatzi, and Eun Pyo Hong
- Subjects
Male ,Huntingtin ,Genome-wide association study ,Disease ,Pathogenesis ,0302 clinical medicine ,Age of Onset ,Aged, 80 and over ,CAG repeat ,Genetics ,Huntingtin Protein ,genetic modifier ,0303 health sciences ,disease modification ,Middle Aged ,Huntington Disease ,Phenotype ,polyglutamine disease ,Female ,age at onset ,trinucleotide repeat ,Huntington’s disease ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA repair ,Biology ,Polymorphism, Single Nucleotide ,Article ,General Biochemistry, Genetics and Molecular Biology ,Young Adult ,03 medical and health sciences ,somatic DNA expansion ,Huntington's disease ,mental disorders ,medicine ,Humans ,Gene ,Alleles ,Aged ,030304 developmental biology ,Base Sequence ,medicine.disease ,nervous system diseases ,Haplotypes ,Genetic Loci ,DNA maintenance ,Peptides ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Summary Variable, glutamine-encoding, CAA interruptions indicate that a property of the uninterrupted HTT CAG repeat sequence, distinct from the length of huntingtin’s polyglutamine segment, dictates the rate at which Huntington’s disease (HD) develops. The timing of onset shows no significant association with HTT cis-eQTLs but is influenced, sometimes in a sex-specific manner, by polymorphic variation at multiple DNA maintenance genes, suggesting that the special onset-determining property of the uninterrupted CAG repeat is a propensity for length instability that leads to its somatic expansion. Additional naturally occurring genetic modifier loci, defined by GWAS, may influence HD pathogenesis through other mechanisms. These findings have profound implications for the pathogenesis of HD and other repeat diseases and question the fundamental premise that polyglutamine length determines the rate of pathogenesis in the “polyglutamine disorders.”, Graphical Abstract, Highlights • Uninterrupted CAG repeat, not polyglutamine, size drives the timing of HD onset • HD age at onset is influenced by at least six genes involved in DNA maintenance • Genetic modifier loci often show both onset-delaying and onset-hastening haplotypes • The rate-determining mechanism is likely to be somatic expansion of the CAG repeat, The onset of Huntington’s disease is shown to be dependent on the size of the uninterrupted CAG repeat sequence, not polyglutamine.
- Published
- 2019
47. Striatal Synaptosomes from Hdh140Q/140Q Knock-in Mice have Altered Protein Levels, Novel Sites of Methionine Oxidation, and Excess Glutamate Release after Stimulation
- Author
-
Scott A. Shaffer, Seung Kwak, Kwadwo A. Ansong, Marian DiFiglia, Ellen Sapp, Antonio Valencia, Hollis McClory, Neil Aronin, Jeffrey S. Kimm, George J. Yohrling, Karin M. Green, and Kimberly B. Kegel
- Subjects
Vesicle fusion ,Blotting, Western ,Glutamic Acid ,Nerve Tissue Proteins ,Stimulation ,Biology ,Article ,Mice ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Methionine ,Huntington's disease ,Tandem Mass Spectrometry ,Neurotransmitter receptor ,medicine ,Animals ,Gene Knock-In Techniques ,Huntingtin Protein ,Vesicle ,Glutamate receptor ,Nuclear Proteins ,Glutamic acid ,medicine.disease ,Corpus Striatum ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Huntington Disease ,chemistry ,Biochemistry ,Neurology (clinical) ,Oxidation-Reduction ,Chromatography, Liquid ,Synaptosomes - Abstract
Synaptic connections are disrupted in patients with Huntington's disease (HD). Synaptosomes from postmortem brain are ideal for synaptic function studies because they are enriched in pre- and post-synaptic proteins important in vesicle fusion, vesicle release, and neurotransmitter receptor activation.To examine striatal synaptosomes from 3, 6 and 12 month old WT and Hdh140Q/140Q knock-in mice for levels of synaptic proteins, methionine oxidation, and glutamate release.We used Western blot analysis, glutamate release assays, and liquid chromatography tandem mass spectrometry (LC-MS/MS).Striatal synaptosomes of 6 month old Hdh140Q/140Q mice had less DARPP32, syntaxin 1 and calmodulin compared to WT. Striatal synaptosomes of 12 month old Hdh140Q/140Q mice had lower levels of DARPP32, alpha actinin, HAP40, Na+/K+-ATPase, PSD95, SNAP-25, TrkA and VAMP1, VGlut1 and VGlut2, increased levels of VAMP2, and modifications in actin and calmodulin compared to WT. More glutamate released from vesicles of depolarized striatal synaptosomes of 6 month old Hdh140Q/140Q than from age matched WT mice but there was no difference in glutamate release in synaptosomes of 3 and 12 month old WT and Hdh140Q/140Q mice. LC-MS/MS of 6 month old Hdh140Q/140Q mice striatal synaptosomes revealed that about 4% of total proteins detected (600 detected) had novel sites of methionine oxidation including proteins involved with vesicle fusion, trafficking, and neurotransmitter function (synaptophysin, synapsin 2, syntaxin 1, calmodulin, cytoplasmic actin 2, neurofilament, and tubulin). Altered protein levels and novel methionine oxidations were also seen in cortical synaptosomes of 12 month old Hdh140Q/140Q mice.Findings provide support for early synaptic dysfunction in Hdh140Q/140Q knock-in mice arising from altered protein levels, oxidative damage, and impaired glutamate neurotransmission and suggest that study of synaptosomes could be of value for evaluating HD therapies.
- Published
- 2013
48. High resolution time-course mapping of early transcriptomic, molecular and cellular phenotypes in Huntington's disease CAG knock-in mice across multiple genetic backgrounds
- Author
-
Andrea Grindeland, David Baxter, Vanessa C. Wheeler, John C. Earls, George A. Carlson, Marina Kovalenko, Nathan D. Price, Leroy Hood, James F. Gusella, Tammy Gillis, Jayalakshmi S. Mysore, Marcy E. MacDonald, Jocelynn R. Pearl, Jeffrey B. Carroll, Kai Wang, Seung Kwak, Jason St. Claire, Min Young Lee, Donald Geman, Jong-Min Lee, Kelsey Scherler, Seth A. Ament, and David Howland
- Subjects
0301 basic medicine ,Huntingtin ,Biology ,medicine.disease_cause ,Genomic Instability ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Huntington's disease ,Gene knockin ,Genetics ,medicine ,Huntingtin Protein ,Animals ,Humans ,Gene Knock-In Techniques ,Allele ,Molecular Biology ,Genetics (clinical) ,Neurons ,Mutation ,Gene Expression Regulation, Developmental ,General Medicine ,Articles ,medicine.disease ,Corpus Striatum ,Disease Models, Animal ,030104 developmental biology ,Huntington Disease ,Phenotype ,Age of onset ,Trinucleotide repeat expansion ,Transcriptome ,Trinucleotide Repeat Expansion ,Genetic Background ,030217 neurology & neurosurgery - 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.
- Published
- 2016
49. Ablation of central nervous system progenitor cells in transgenic rats using bacterial nitroreductase system
- Author
-
Yin She, David Howland, Afshin Ghavami, Seung Kwak, Ke-Yi Cheng, Myles Fennell, Jessica E. Malberg, and Jianying Su
- Subjects
Central Nervous System ,Programmed cell death ,Cerebellum ,Time Factors ,Aziridines ,Green Fluorescent Proteins ,Population ,Antineoplastic Agents ,Nerve Tissue Proteins ,Biology ,Green fluorescent protein ,Animals, Genetically Modified ,Nestin ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,Intermediate Filament Proteins ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Progenitor cell ,education ,Injections, Intraventricular ,Electroshock ,education.field_of_study ,Behavior, Animal ,Dose-Response Relationship, Drug ,Escherichia coli Proteins ,Stem Cells ,Nitroreductases ,Molecular biology ,Rats ,medicine.anatomical_structure ,Animals, Newborn ,Bromodeoxyuridine ,Cell culture ,Stem cell - Abstract
Specific ablation of central nervous system (CNS) progenitor cells in the brain of live animals is a powerful method to determine the functions of these cells and to reveal novel avenues for the treatment of several CNS-related disorders. To achieve this goal, we generated a line of transgenic rats expressing a bacterial enzyme, Escherichia coli nitroreductase gene (NTR), under control of the nestin promoter. In this system, NTR(+) cells are selectively eliminated upon application of prodrug CB1954, through activation of programmed cell death machineries. At 5 days of age, which is a time when cerebellar development is occurring, transgenic rats bearing the nestin-NTR/green fluorescent protein (GFP) gene are overtly normal and express NTR/GFP in neuronal stem cells, without any toxicity in these cells. The functional consequence of progenitor cell ablation was demonstrated by administering prodrug CB1954 into the cerebellum at this 5-day time point. Stem cell ablation in these neonates resulted in sensorimotor abnormalities, cerebellar degeneration, overall reduction in cerebellar seize, and manifestation of ataxia. In adult rats, GFP expression was not seen in the hippocampal progenitor cells and seen only at very low levels in the lateral ventricles, indicating a different NTR/GFP expression pattern between neonates and adults. In addition, application of CB1954 by intraventricular delivery reduced the number of 5-bromo-2'-deoxyuridine-labeled proliferating cells in the lateral ventricle but not hippocampus of NTR/GFP rats. These findings shows that targeted expression of NTR under a specific promoter might be of significant value in addressing the function of distinct cell population in vivo.
- Published
- 2007
50. Integrated genomics and proteomics define huntingtin CAG length-dependent networks in mice
- Author
-
Fuying Gao, Giovanni Coppola, Sandeep Deverasetty, Andreas Tebbe, Xiao-Hong Lu, Christoph Schaab, Peter Langfelder, X. William Yang, Eliana Marisa Ramos, Nan Wang, Jeffrey P. Cantle, Juan Botas, Ismael Al-Ramahi, Daniel J. Lavery, Jeffrey S. Aaronson, Doxa Chatzopoulou, Yining Zhao, Jim Rosinski, David Howland, Karla El-Zein, Steve Horvath, and Seung Kwak
- Subjects
0301 basic medicine ,Huntington's Disease ,Male ,Proteomics ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Protocadherin ,Nerve Tissue Proteins ,Biology ,Neurodegenerative ,Medium spiny neuron ,Inbred C57BL ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Rare Diseases ,Huntington's disease ,Gene expression ,mental disorders ,Huntingtin Protein ,medicine ,Genetics ,Psychology ,Animals ,Gene Regulatory Networks ,Gene Knock-In Techniques ,Cerebral Cortex ,Neurology & Neurosurgery ,General Neuroscience ,Neurosciences ,Nuclear Proteins ,Genomics ,medicine.disease ,Molecular biology ,Corpus Striatum ,Brain Disorders ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Cerebral cortex ,Neurological ,Cognitive Sciences ,Female ,030217 neurology & neurosurgery ,Biotechnology - Abstract
To gain insight into how mutant huntingtin (mHtt) CAG repeat length modifies Huntington’s disease (HD) pathogenesis, we profiled mRNA in over 600 brain and peripheral tissue samples from HD knock-in mice with increasing CAG repeat lengths. We find repeat length dependent transcriptional signatures are prominent in the striatum, less so in cortex, and minimal in the liver. Co-expression network analyses reveal 13 striatal and 5 cortical modules that are highly correlated with CAG length and age, and that are preserved in HD models and some in the patients. Top striatal modules implicate mHtt CAG length and age in graded impairment of striatal medium spiny neuron identity gene expression and in dysregulation of cAMP signaling, cell death, and protocadherin genes. Importantly, we used proteomics to confirm 790 genes and 5 striatal modules with CAG length-dependent dysregulation at both RNA and protein levels, and validated 22 striatal module genes as modifiers of mHtt toxicities in vivo.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.