143 results on '"Wheeler, Vanessa"'
Search Results
102. Msh2 acts in medium-spiny striatal neurons as an enhancer of CAG instability and mutant huntingtin phenotypes in Huntington's disease knock-in mice.
- Author
-
Kovalenko, Marina, Dragileva, Ella, St Claire, Jason, Gillis, Tammy, Guide, Jolene R, New, Jaclyn, Dong, Hualing, Kucherlapati, Raju, Kucherlapati, Melanie H, Ehrlich, Michelle E, Lee, Jong-Min, Wheeler, Vanessa C, Kovalenko, Marina, Dragileva, Ella, St Claire, Jason, Gillis, Tammy, Guide, Jolene R, New, Jaclyn, Dong, Hualing, Kucherlapati, Raju, Kucherlapati, Melanie H, Ehrlich, Michelle E, Lee, Jong-Min, and Wheeler, Vanessa C
- Abstract
The CAG trinucleotide repeat mutation in the Huntington's disease gene (HTT) exhibits age-dependent tissue-specific expansion that correlates with disease onset in patients, implicating somatic expansion as a disease modifier and potential therapeutic target. Somatic HTT CAG expansion is critically dependent on proteins in the mismatch repair (MMR) pathway. To gain further insight into mechanisms of somatic expansion and the relationship of somatic expansion to the disease process in selectively vulnerable MSNs we have crossed HTT CAG knock-in mice (HdhQ111) with mice carrying a conditional (floxed) Msh2 allele and D9-Cre transgenic mice, in which Cre recombinase is expressed specifically in MSNs within the striatum. Deletion of Msh2 in MSNs eliminated Msh2 protein in those neurons. We demonstrate that MSN-specific deletion of Msh2 was sufficient to eliminate the vast majority of striatal HTT CAG expansions in HdhQ111 mice. Furthermore, MSN-specific deletion of Msh2 modified two mutant huntingtin phenotypes: the early nuclear localization of diffusely immunostaining mutant huntingtin was slowed; and the later development of intranuclear huntingtin inclusions was dramatically inhibited. Therefore, Msh2 acts within MSNs as a genetic enhancer both of somatic HTT CAG expansions and of HTT CAG-dependent phenotypes in mice. These data suggest that the selective vulnerability of MSNs may be at least in part contributed by the propensity for somatic expansion in these neurons, and imply that intervening in the expansion process is likely to have therapeutic benefit.
- Published
- 2012
103. A Broad Phenotypic Screen Identifies Novel Phenotypes Driven by a Single Mutant Allele in Huntington’s Disease CAG Knock-In Mice
- Author
-
Hölter, Sabine M., primary, Stromberg, Mary, additional, Kovalenko, Marina, additional, Garrett, Lillian, additional, Glasl, Lisa, additional, Lopez, Edith, additional, Guide, Jolene, additional, Götz, Alexander, additional, Hans, Wolfgang, additional, Becker, Lore, additional, Rathkolb, Birgit, additional, Rozman, Jan, additional, Schrewed, Anja, additional, Klingenspor, Martin, additional, Klopstock, Thomas, additional, Schulz, Holger, additional, Wolf, Eckhard, additional, Wursta, Wolfgang, additional, Gillis, Tammy, additional, Wakimoto, Hiroko, additional, Seidman, Jonathan, additional, MacDonald, Marcy E., additional, Cotman, Susan, additional, Gailus-Durner, Valérie, additional, Fuchs, Helmut, additional, de Angelis, Martin Hrabě, additional, Lee, Jong-Min, additional, and Wheeler, Vanessa C., additional
- Published
- 2013
- Full Text
- View/download PDF
104. Mismatch Repair Genes Mlh1 and Mlh3 Modify CAG Instability in Huntington's Disease Mice: Genome-Wide and Candidate Approaches
- Author
-
Pinto, Ricardo Mouro, primary, Dragileva, Ella, additional, Kirby, Andrew, additional, Lloret, Alejandro, additional, Lopez, Edith, additional, St. Claire, Jason, additional, Panigrahi, Gagan B., additional, Hou, Caixia, additional, Holloway, Kim, additional, Gillis, Tammy, additional, Guide, Jolene R., additional, Cohen, Paula E., additional, Li, Guo-Min, additional, Pearson, Christopher E., additional, Daly, Mark J., additional, and Wheeler, Vanessa C., additional
- Published
- 2013
- Full Text
- View/download PDF
105. Large-scale phenome analysis defines a behavioral signature for Huntington's disease genotype in mice.
- Author
-
Alexandrov, Vadim, Brunner, Dani, Menalled, Liliana B, Kudwa, Andrea, Watson-Johnson, Judy, Mazzella, Matthew, Russell, Ian, Ruiz, Melinda C, Torello, Justin, Sabath, Emily, Sanchez, Ana, Gomez, Miguel, Filipov, Igor, Cox, Kimberly, Kwan, Mei, Ghavami, Afshin, Ramboz, Sylvie, Lager, Brenda, Wheeler, Vanessa C, and Aaronson, Jeff
- Published
- 2016
- Full Text
- View/download PDF
106. Msh2 Acts in Medium-Spiny Striatal Neurons as an Enhancer of CAG Instability and Mutant Huntingtin Phenotypes in Huntington’s Disease Knock-In Mice
- Author
-
Kovalenko, Marina, primary, Dragileva, Ella, additional, St. Claire, Jason, additional, Gillis, Tammy, additional, Guide, Jolene R., additional, New, Jaclyn, additional, Dong, Hualing, additional, Kucherlapati, Raju, additional, Kucherlapati, Melanie H., additional, Ehrlich, Michelle E., additional, Lee, Jong-Min, additional, and Wheeler, Vanessa C., additional
- Published
- 2012
- Full Text
- View/download PDF
107. Large-Scale Phenotyping of an Accurate Genetic Mouse Model of JNCL Identifies Novel Early Pathology Outside the Central Nervous System
- Author
-
Staropoli, John F., primary, Haliw, Larissa, additional, Biswas, Sunita, additional, Garrett, Lillian, additional, Hölter, Sabine M., additional, Becker, Lore, additional, Skosyrski, Sergej, additional, Da Silva-Buttkus, Patricia, additional, Calzada-Wack, Julia, additional, Neff, Frauke, additional, Rathkolb, Birgit, additional, Rozman, Jan, additional, Schrewe, Anja, additional, Adler, Thure, additional, Puk, Oliver, additional, Sun, Minxuan, additional, Favor, Jack, additional, Racz, Ildikó, additional, Bekeredjian, Raffi, additional, Busch, Dirk H., additional, Graw, Jochen, additional, Klingenspor, Martin, additional, Klopstock, Thomas, additional, Wolf, Eckhard, additional, Wurst, Wolfgang, additional, Zimmer, Andreas, additional, Lopez, Edith, additional, Harati, Hayat, additional, Hill, Eric, additional, Krause, Daniela S., additional, Guide, Jolene, additional, Dragileva, Ella, additional, Gale, Evan, additional, Wheeler, Vanessa C., additional, Boustany, Rose-Mary, additional, Brown, Diane E., additional, Breton, Sylvie, additional, Ruether, Klaus, additional, Gailus-Durner, Valérie, additional, Fuchs, Helmut, additional, de Angelis, Martin Hrabě, additional, and Cotman, Susan L., additional
- Published
- 2012
- Full Text
- View/download PDF
108. Quantification of Age-Dependent Somatic CAG Repeat Instability in Hdh CAG Knock-In Mice Reveals Different Expansion Dynamics in Striatum and Liver
- Author
-
Lee, Jong-Min, primary, Pinto, Ricardo Mouro, additional, Gillis, Tammy, additional, St. Claire, Jason C., additional, and Wheeler, Vanessa C., additional
- Published
- 2011
- Full Text
- View/download PDF
109. Creation of an Open-Access, Mutation-Defined Fibroblast Resource for Neurological Disease Research
- Author
-
Wray, S, Self, M, NINDS Parkinson's Disease iPSC Consortium, Gusella, NINDS Huntington's Disease iPSC Consortium James F., Macdonald, Marcy E., Wheeler, Vanessa C., Ross, Christopher A., Sergey, Akimov, Jamshid, Arjomand, Thompson, Leslie M., Alvin, King, Neal, Hermanowicz, Sara, Winokur, Svendsen, Clive N., Virginia, Mattis, Onorati, Marco, Elena, Cattaneo, Allen, Nicholas D., Kemp, Paul J., Kwang Soo Kim, Steven, Finkbeiner, NINDS ALS iPSC Consortium, Lewis, Pa, Taanman, Jw, Ryan, Ns, Mahoney, Cj, Liang, Y, Devine, Mj, Sheerin, Um, Houlden, H, Morris, Hr, Healy, D, Marti Masso JF, Preza, E, Barker, S, Sutherland, M, Corriveau, Ra, D'Andrea, M, Schapira, Ah, Uitti, Rj, Guttman, M, Opala, G, Jasinska Myga, B, Puschmann, A, Nilsson, C, Espay, Aj, Slawek, J, Gutmann, L, Boeve, Bf, Boylan, K, Stoessl, Aj, Ross, Oa, Maragakis, Nj, Van Gerpen, J, Gerstenhaber, M, Gwinn, K, Dawson, Tm, Isacson, O, Marder, Ks, Clark, Ln, Przedborski, Se, Finkbeiner, S, Rothstein, Jd, Wszolek, Zk, Rossor, Mn, Hardy, J., and Borlongan, Cesar V
- Subjects
Neurology ,Databases, Factual ,Biopsy ,Cellular differentiation ,lcsh:Medicine ,Disease ,Bioinformatics ,Motor Neuron Diseases ,0302 clinical medicine ,Models ,Neurobiology of Disease and Regeneration ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,NINDS ALS iPSC Consortium ,Induced pluripotent stem cell ,Psychiatry ,0303 health sciences ,Multidisciplinary ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Molecular pathology ,Parkinson Disease ,Cell Differentiation ,Medical research ,Immunohistochemistry ,3. Good health ,NINDS Parkinson's Disease iPSC Consortium ,Autosomal Dominant ,Neurological ,Medicine ,Medical genetics ,Alzheimer's disease ,Research Article ,medicine.medical_specialty ,General Science & Technology ,Induced Pluripotent Stem Cells ,Tissue Banks ,Cell Line ,Access to Information ,Databases ,03 medical and health sciences ,Genetic ,Alzheimer Disease ,Genetics ,medicine ,Humans ,QH426 ,Biology ,Factual ,Cell Proliferation ,030304 developmental biology ,Clinical Genetics ,Stem Cell Research - Induced Pluripotent Stem Cell ,Models, Genetic ,business.industry ,lcsh:R ,Neurosciences ,Fibroblasts ,Stem Cell Research ,medicine.disease ,R1 ,Brain Disorders ,NINDS Huntington's Disease iPSC Consortium ,Geriatrics ,Mutation ,lcsh:Q ,Dementia ,Generic health relevance ,Molecular Neuroscience ,Nervous System Diseases ,business ,2.6 Resources and infrastructure (aetiology) ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Our understanding of the molecular mechanisms of many neurological disorders has been greatly enhanced by the discovery of mutations in genes linked to familial forms of these diseases. These have facilitated the generation of cell and animal models that can be used to understand the underlying molecular pathology. Recently, there has been a surge of interest in the use of patient-derived cells, due to the development of induced pluripotent stem cells and their subsequent differentiation into neurons and glia. Access to patient cell lines carrying the relevant mutations is a limiting factor for many centres wishing to pursue this research. We have therefore generated an open-access collection of fibroblast lines from patients carrying mutations linked to neurological disease. These cell lines have been deposited in the National Institute for Neurological Disorders and Stroke (NINDS) Repository at the Coriell Institute for Medical Research and can be requested by any research group for use in in vitro disease modelling. There are currently 71 mutation-defined cell lines available for request from a wide range of neurological disorders and this collection will be continually expanded. This represents a significant resource that will advance the use of patient cells as disease models by the scientific community.
- Published
- 2012
- Full Text
- View/download PDF
110. A novel approach to investigate tissue-specific trinucleotide repeat instability
- Author
-
Lee, Jong-Min, primary, Zhang, Jie, additional, Su, Andrew I, additional, Walker, John R, additional, Wiltshire, Tim, additional, Kang, Kihwa, additional, Dragileva, Ella, additional, Gillis, Tammy, additional, Lopez, Edith T, additional, Boily, Marie-Josee, additional, Cyr, Michel, additional, Kohane, Isaac, additional, Gusella, James F, additional, MacDonald, Marcy E, additional, and Wheeler, Vanessa C, additional
- Published
- 2010
- Full Text
- View/download PDF
111. Huntingtin facilitates polycomb repressive complex 2
- Author
-
Seong, Ihn Sik, primary, Woda, Juliana M., additional, Song, Ji-Joon, additional, Lloret, Alejandro, additional, Abeyrathne, Priyanka D., additional, Woo, Caroline J., additional, Gregory, Gillian, additional, Lee, Jong-Min, additional, Wheeler, Vanessa C., additional, Walz, Thomas, additional, Kingston, Robert E., additional, Gusella, James F., additional, Conlon, Ronald A., additional, and MacDonald, Marcy E., additional
- Published
- 2009
- Full Text
- View/download PDF
112. The Genetic Modifiers of Motor Onset Age (GeM MOA) Website: Genome-wide Association Analysis for Genetic Modifiers of Huntington's Disease.
- Author
-
Correia, Kevin, Harold, Denise, Kyung-Hee Kim, Holmans, Peter, Jones, Lesley, Orth, Michael, Myers, Richard H., Seung Kwak, Wheeler, Vanessa C., MacDonald, Marcy E., Gusella, James F., and Jong-Min Lee
- Subjects
HUNTINGTON disease ,IMMUNOMODULATORS ,NEURODEGENERATION ,TRINUCLEOTIDE repeats ,HUMAN genome - Abstract
Background: Huntington's disease (HD) is a dominantly inherited disease caused by a CAG expansion mutation in HTT. The age at onset of clinical symptoms is determined primarily by the length of this CAG expansion but is also influenced by other genetic and/or environmental factors. Objective: Recently, through genome-wide association studies (GWAS) aimed at discovering genetic modifiers, we identified loci associated with age at onset of motor signs that are significant at the genome-wide level. However, many additional HD modifiers may exist but may not have achieved statistical significance due to limited power. Methods: In order to disseminate broadly the entire GWAS results and make them available to complement alternative approaches, we have developed the internet website "GeM MOA" where genetic association results can be searched by gene name, SNP ID, or genomic coordinates of a region of interest. Results: Users of the Genetic Modifiers of Motor Onset Age (GeM MOA) site can therefore examine support for association between any gene region and age at onset of HD motor signs. GeM MOA's interactive interface also allows users to navigate the surrounding region and to obtain association p-values for individual SNPs. Conclusions: Our website conveys a comprehensive view of the genetic landscape of modifiers of HD from the existing GWAS, and will provide the means to evaluate the potential influence of genes of interest on the onset of HD. GeM MOA is freely available at https://www.hdinhd.org/. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
113. Genome-wide significance for a modifier of age at neurological onset in Huntington's Disease at 6q23-24: the HD MAPS study
- Author
-
Li, Jian-Liang, primary, Hayden, Michael R, additional, Warby, Simon C, additional, Durr, Alexandra, additional, Morrison, Patrick J, additional, Nance, Martha, additional, Ross, Christopher A, additional, Margolis, Russell L, additional, Rosenblatt, Adam, additional, Squitieri, Ferdinando, additional, Frati, Luigi, additional, Gómez-Tortosa, Estrella, additional, García, Carmen Ayuso, additional, Suchowersky, Oksana, additional, Klimek, Mary Lou, additional, Trent, Ronald JA, additional, McCusker, Elizabeth, additional, Novelletto, Andrea, additional, Frontali, Marina, additional, Paulsen, Jane S, additional, Jones, Randi, additional, Ashizawa, Tetsuo, additional, Lazzarini, Alice, additional, Wheeler, Vanessa C, additional, Prakash, Ranjana, additional, Xu, Gang, additional, Djoussé, Luc, additional, Mysore, Jayalakshmi Srinidhi, additional, Gillis, Tammy, additional, Hakky, Michael, additional, Cupples, L Adrienne, additional, Saint-Hilaire, Marie H, additional, Cha, Jang-Ho J, additional, Hersch, Steven M, additional, Penney, John B, additional, Harrison, Madaline B, additional, Perlman, Susan L, additional, Zanko, Andrea, additional, Abramson, Ruth K, additional, Lechich, Anthony J, additional, Duckett, Ayana, additional, Marder, Karen, additional, Conneally, P Michael, additional, Gusella, James F, additional, MacDonald, Marcy E, additional, and Myers, Richard H, additional
- Published
- 2006
- Full Text
- View/download PDF
114. Genetic Contributors to Intergenerational CAG Repeat Instability in Huntington’s Disease Knock-In Mice
- Author
-
Neto, João Luís, Lee, Jong-Min, Afridi, Ali, Gillis, Tammy, Guide, Jolene R., Dempsey, Stephani, Lager, Brenda, Alonso, Isabel, Wheeler, Vanessa C., and Pinto, Ricardo Mouro
- Subjects
Genome Integrity and Transmission ,Huntington’s disease ,intergenerational CAG repeat instability ,HD knock-in mouse models ,genetic background - Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by the expansion of a CAG trinucleotide repeat in exon 1 of the HTT gene. Longer repeat sizes are associated with increased disease penetrance and earlier ages of onset. Intergenerationally unstable transmissions are common in HD families, partly underlying the genetic anticipation seen in this disorder. HD CAG knock-in mouse models also exhibit a propensity for intergenerational repeat size changes. In this work, we examine intergenerational instability of the CAG repeat in over 20,000 transmissions in the largest HD knock-in mouse model breeding datasets reported to date. We confirmed previous observations that parental sex drives the relative ratio of expansions and contractions. The large datasets further allowed us to distinguish effects of paternal CAG repeat length on the magnitude and frequency of expansions and contractions, as well as the identification of large repeat size jumps in the knock-in models. Distinct degrees of intergenerational instability were observed between knock-in mice of six background strains, indicating the occurrence of trans-acting genetic modifiers. We also found that lines harboring a neomycin resistance cassette upstream of Htt showed reduced expansion frequency, indicative of a contributing role for sequences in cis, with the expanded repeat as modifiers of intergenerational instability. These results provide a basis for further understanding of the mechanisms underlying intergenerational repeat instability.
- Published
- 2017
- Full Text
- View/download PDF
115. The HD Mutation Does Not Alter Neuronal Death in the Striatum of HdhQ92 Knock-in Mice after Mild Focal Ischemia
- Author
-
Namura, Shobu, primary, Hirt, Lorenz, additional, Wheeler, Vanessa C., additional, McGinnis, Kim M., additional, Hilditch-Maguire, Paige, additional, Moskowitz, Michael A., additional, MacDonald, Marcy E., additional, and Persichetti, Francesca, additional
- Published
- 2002
- Full Text
- View/download PDF
116. Rolling Circle Amplification
- Author
-
Gusev, Yuriy, primary, Sparkowski, Jason, additional, Raghunathan, Arumugham, additional, Ferguson, Harley, additional, Montano, Jane, additional, Bogdan, Nancy, additional, Schweitzer, Barry, additional, Wiltshire, Steven, additional, Kingsmore, Stephen F., additional, Maltzman, Warren, additional, and Wheeler, Vanessa, additional
- Published
- 2001
- Full Text
- View/download PDF
117. Modification of the mouse mitochondrial genome by insertion of an exogenous gene
- Author
-
Wheeler, Vanessa C., primary, Aitken, Mark, additional, and Coutelle, Charles, additional
- Published
- 1997
- Full Text
- View/download PDF
118. Introduction of Plasmid DNA into Isolated Mitochondria by Electroporation
- Author
-
Collombet, Jean-Marc, primary, Wheeler, Vanessa C., additional, Vogel, Frank, additional, and Coutelle, Charles, additional
- Published
- 1997
- Full Text
- View/download PDF
119. Synthesis of a modified gene encoding human ornithine transcarbamylase for expression in mammalian mitochondrial and universal translation systems: a novel approach towards correction of a genetic defect
- Author
-
Wheeler, Vanessa C., primary, Prodromou, Chrisostomos, additional, Pearl, Laurence H., additional, Williamson, Robert, additional, and Coutelle, Charles, additional
- Published
- 1996
- Full Text
- View/download PDF
120. Mismatch Repair Genes Mlh1 and Mlh3 Modify CAG Instability in Huntington's Disease Mice: Genome-Wide and Candidate Approaches.
- Author
-
Pinto, Ricardo Mouro, Dragileva, Ella, Kirby, Andrew, Lloret, Alejandro, Lopez, Edith, St. Claire, Jason, Panigrahi, Gagan B., Hou, Caixia, Holloway, Kim, Gillis, Tammy, Guide, Jolene R., Cohen, Paula E., Li, Guo-Min, Pearson, Christopher E., Daly, Mark J., and Wheeler, Vanessa C.
- Subjects
DNA repair ,HUNTINGTON disease ,LABORATORY mice ,MMR vaccines ,GENETIC mutation ,MLH1 gene ,GENETICS - Abstract
The Huntington's disease gene (HTT) CAG repeat mutation undergoes somatic expansion that correlates with pathogenesis. Modifiers of somatic expansion may therefore provide routes for therapies targeting the underlying mutation, an approach that is likely applicable to other trinucleotide repeat diseases. Huntington's disease Hdh
Q111 mice exhibit higher levels of somatic HTT CAG expansion on a C57BL/6 genetic background (B6.HdhQ111 ) than on a 129 background (129.HdhQ111 ). Linkage mapping in (B6x129).HdhQ111 F2 intercross animals identified a single quantitative trait locus underlying the strain-specific difference in expansion in the striatum, implicating mismatch repair (MMR) gene Mlh1 as the most likely candidate modifier. Crossing B6.HdhQ111 mice onto an Mlh1 null background demonstrated that Mlh1 is essential for somatic CAG expansions and that it is an enhancer of nuclear huntingtin accumulation in striatal neurons. HdhQ111 somatic expansion was also abolished in mice deficient in the Mlh3 gene, implicating MutLγ (MLH1–MLH3) complex as a key driver of somatic expansion. Strikingly, Mlh1 and Mlh3 genes encoding MMR effector proteins were as critical to somatic expansion as Msh2 and Msh3 genes encoding DNA mismatch recognition complex MutSβ (MSH2–MSH3). The Mlh1 locus is highly polymorphic between B6 and 129 strains. While we were unable to detect any difference in base-base mismatch or short slipped-repeat repair activity between B6 and 129 MLH1 variants, repair efficiency was MLH1 dose-dependent. MLH1 mRNA and protein levels were significantly decreased in 129 mice compared to B6 mice, consistent with a dose-sensitive MLH1-dependent DNA repair mechanism underlying the somatic expansion difference between these strains. Together, these data identify Mlh1 and Mlh3 as novel critical genetic modifiers of HTT CAG instability, point to Mlh1 genetic variation as the likely source of the instability difference in B6 and 129 strains and suggest that MLH1 protein levels play an important role in driving of the efficiency of somatic expansions. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
121. Msh2Acts in Medium-Spiny Striatal Neurons as an Enhancer of CAG Instability and Mutant Huntingtin Phenotypes in Huntington's Disease Knock-In Mice.
- Author
-
Kovalenko, Marina, Dragileva, Ella, St. Claire, Jason, Gillis, Tammy, Guide, Jolene R., New, Jaclyn, Hualing Dong, Kucherlapati, Raju, Kucherlapati, Melanie H., Ehrlich, Michelle E., Jong-Min Lee, Wheeler, Vanessa C., and Hoyt, Kari
- Subjects
NEURONS ,TRINUCLEOTIDE repeats ,HUNTINGTON disease ,TISSUE-specific antibodies ,LABORATORY mice ,MOUSE diseases - Abstract
The CAG trinucleotide repeat mutation in the Huntington's disease gene (HTT) exhibits age-dependent tissue-specific expansion that correlates with disease onset in patients, implicating somatic expansion as a disease modifier and potential therapeutic target. Somatic HTT CAG expansion is critically dependent on proteins in the mismatch repair (MMR) pathway. To gain further insight into mechanisms of somatic expansion and the relationship of somatic expansion to the disease process in selectively vulnerable MSNs we have crossed HTT CAG knock-in mice (HdhQ111) with mice carrying a conditional (floxed) Msh2 allele and D9-Cre transgenic mice, in which Cre recombinase is expressed specifically in MSNs within the striatum. Deletion of Msh2 in MSNs eliminated Msh2 protein in those neurons. We demonstrate that MSN-specific deletion of Msh2 was sufficient to eliminate the vast majority of striatal HTT CAG expansions in HdhQ111 mice. Furthermore, MSNspecific deletion of Msh2 modified two mutant huntingtin phenotypes: the early nuclear localization of diffusely immunostaining mutant huntingtin was slowed; and the later development of intranuclear huntingtin inclusions was dramatically inhibited. Therefore, Msh2 acts within MSNs as a genetic enhancer both of somatic HTT CAG expansions and of HTT CAG-dependent phenotypes in mice. These data suggest that the selective vulnerability of MSNs may be at least in part contributed by the propensity for somatic expansion in these neurons, and imply that intervening in the expansion process is likely to have therapeutic benefit. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
122. HA novel approach to investigate tissue-specific trinucleotide repeat instability.
- Author
-
Jong-Min Lee, Jie Zhang, Su, Andrew I., Walker, John R., Wiltshire, Tim, Kihwa Kang, Dragileva, Ella, Gillis, Tammy, Lopez, Edith T., Boily, Marie-Josee, Cyr, Michel, Kohane, Isaac, Gusella, James F., MacDonald, Marcy E., and Wheeler, Vanessa C.
- Subjects
HUNTINGTON disease ,NEURODEGENERATION ,GENOMES ,BIOINFORMATICS ,GENE expression - Abstract
Background: In Huntington's disease (HD), an expanded CAG repeat produces characteristic striatal neurodegeneration. Interestingly, the HD CAG repeat, whose length determines age at onset, undergoes tissuespecific somatic instability, predominant in the striatum, suggesting that tissue-specific CAG length changes could modify the disease process. Therefore, understanding the mechanisms underlying the tissue specificity of somatic instability may provide novel routes to therapies. However progress in this area has been hampered by the lack of sensitive high-throughput instability quantification methods and global approaches to identify the underlying factors. Results: Here we describe a novel approach to gain insight into the factors responsible for the tissue specificity of somatic instability. Using accurate genetic knock-in mouse models of HD, we developed a reliable, highthroughput method to quantify tissue HD CAG repeat instability and integrated this with genome-wide bioinformatic approaches. Using tissue instability quantified in 16 tissues as a phenotype and tissue microarray gene expression as a predictor, we built a mathematical model and identified a gene expression signature that accurately predicted tissue instability. Using the predictive ability of this signature we found that somatic instability was not a consequence of pathogenesis. In support of this, genetic crosses with models of accelerated neuropathology failed to induce somatic instability. In addition, we searched for genes and pathways that correlated with tissue instability. We found that expression levels of DNA repair genes did not explain the tissue specificity of somatic instability. Instead, our data implicate other pathways, particularly cell cycle, metabolism and neurotransmitter pathways, acting in combination to generate tissue-specific patterns of instability. Conclusion: Our study clearly demonstrates that multiple tissue factors reflect the level of somatic instability in different tissues. In addition, our quantitative, genome-wide approach is readily applicable to high-throughput assays and opens the door to widespread applications with the potential to accelerate the discovery of drugs that alter tissue instability. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
123. Stoichiometry of Base Excision Repair Proteins Correlates with Increased Somatic CAG Instability in Striatum over Cerebellum in Huntington's Disease Transgenic Mice.
- Author
-
Agathi-Vassiliki Goula, Berquist, Brian R., Wilson III, David M., Wheeler, Vanessa C., Trottier, Yvon, and Merienne, Karine
- Subjects
HUNTINGTON disease ,STOICHIOMETRY ,CEREBELLUM ,TRANSGENIC mice ,GERM cells ,SOMATIC cells ,DNA damage - Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by expansion of an unstable CAG repeat in the coding sequence of the Huntingtin (HTT) gene. Instability affects both germline and somatic cells. Somatic instability increases with age and is tissue-specific. In particular, the CAG repeat sequence in the striatum, the brain region that preferentially degenerates in HD, is highly unstable, whereas it is rather stable in the disease-spared cerebellum. The mechanisms underlying the age-dependence and tissue-specificity of somatic CAG instability remain obscure. Recent studies have suggested that DNA oxidation and OGG1, a glycosylase involved in the repair of 8-oxoguanine lesions, contribute to this process. We show that in HD mice oxidative DNA damage abnormally accumulates at CAG repeats in a length-dependent, but age- and tissue-independent manner, indicating that oxidative DNA damage alone is not sufficient to trigger somatic instability. Protein levels and activities of major base excision repair (BER) enzymes were compared between striatum and cerebellum of HD mice. Strikingly, 5′-flap endonuclease activity was much lower in the striatum than in the cerebellum of HD mice. Accordingly, Flap Endonuclease-1 (FEN1), the main enzyme responsible for 5′-flap endonuclease activity, and the BER cofactor HMGB1, both of which participate in long-patch BER (LP-BER), were also significantly lower in the striatum compared to the cerebellum. Finally, chromatin immunoprecipitation experiments revealed that POLβ was specifically enriched at CAG expansions in the striatum, but not in the cerebellum of HD mice. These in vivo data fit a model in which POLβ strand displacement activity during LP-BER promotes the formation of stable 5′-flap structures at CAG repeats representing pre-expanded intermediate structures, which are not efficiently removed when FEN1 activity is constitutively low. We propose that the stoichiometry of BER enzymes is one critical factor underlying the tissue selectivity of somatic CAG expansion. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
124. The HD mutation causes progressivelethal neurological disease in mice expressing reduced levels ofhuntingtin.
- Author
-
Auerbach, Wojtek, Hurlbert, MarcS., Hilditch-Maguire, Paige, Wadghiri, Youssef Zaim, Wheeler, Vanessa C., Cohen, Sara I., Joyner, AlexandraL., MacDonald, MarcyE., and Turnbull, DanielH.
- Published
- 2001
- Full Text
- View/download PDF
125. Length-dependent gametic CAG repeat instability in the Huntington's disease knock-in mouse.
- Author
-
Wheeler, Vanessa C., Auerbach, Wojtek, White, Jacqueline K., Srinidhi, Jayalakshmi, Auerbach, Anna, Ryan, Angela, Duyao, Mabel P., Vrbanac, Vladimir, Weaver, Meredith, Gusella, James F., Joyner, Alexandra L., and MacDonald, Marcy E.
- Abstract
The CAG repeats in the human Huntington's disease (HD) gene exhibit striking length-dependent intergenerational instability, typically small size increases or decreases of one to a few CAGs, but little variation in somatic tissues. In a subset of male transmissions, larger size increases occur to produce extreme HD alleles that display somatic instability and cause juvenile onset of the disorder. Initial efforts to reproduce these features in a mouse model transgenic for HD exon 1 with 48 CAG repeats revealed only mild intergenerational instability (∼2% of meioses). A similar pattern was obtained when this repeat was inserted into exon 1 of the mouse Hdh gene. However, lengthening the repeats in Hdh to 90 and 109 units produced a graded increase in the mutation frequency to >70%, with instability being more evident in female transmissions. No large jumps in CAG length were detected in either male or female transmissions. Instead, size changes were modest increases and decreases, with expansions typically emanating from males and contractions from females. Limited CAG variation in the somatic tissues gave way to marked mosaicism in liver and striatum for the longest repeats in older mice. These results indicate that gametogenesis is the primary source of inherited instability in the Hdh knock-in mouse, as it is in man, but that the underlying repeat length-dependent mechanism, which may or may not be related in the two species, operates at higher CAG numbers. Moreover, the large CAG repeat increases seen in a subset of male HD transmissions are not reproduced in the mouse, suggesting that these arise by a different fundamental mechanism than the small size fluctuations that are frequent during gametogenesis in both species. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
126. Editorial.
- Author
-
Leavitt, Blair R., Thompson, Leslie M., Jones, Lesley, Pearson, Christopher E., and Wheeler, Vanessa
- Subjects
HUNTINGTON disease ,GENETIC testing ,MYOTONIA atrophica - Published
- 2021
- Full Text
- View/download PDF
127. Huntingtin Facilitates Polycomb Repressive Complex 2
- Author
-
Woda, Juliana M., Song, Ji-Joon, Lloret, Alejandro, Abeyrathne, Priyanka D., Gregory, Gillian, Lee, Jong-Min, Conlon, Ronald A., Seong, Ihn Sik, Woo, Caroline, Wheeler, Vanessa Chantal, Walz, Thomas, Kingston, Robert Edward, Gusella, James Francis, and MacDonald, Marcy Elizabeth
- Abstract
Huntington's disease (HD) is caused by expansion of the polymorphic polyglutamine segment in the huntingtin protein. Full-length huntingtin is thought to be a predominant HEAT repeat α-solenoid, implying a role as a facilitator of macromolecular complexes. Here we have investigated huntingtin's domain structure and potential intersection with epigenetic silencer polycomb repressive complex 2 (PRC2), suggested by shared embryonic deficiency phenotypes. Analysis of a set of full-length recombinant huntingtins, with different polyglutamine regions, demonstrated dramatic conformational flexibility, with an accessible hinge separating two large α-helical domains. Moreover, embryos lacking huntingtin exhibited impaired PRC2 regulation of Hox gene expression, trophoblast giant cell differentiation, paternal X chromosome inactivation and histone H3K27 tri-methylation, while full-length endogenous nuclear huntingtin in wild-type embryoid bodies (EBs) was associated with PRC2 subunits and was detected with trimethylated histone H3K27 at Hoxb9. Supporting a direct stimulatory role, full-length recombinant huntingtin significantly increased the histone H3K27 tri-methylase activity of reconstituted PRC2 in vitro, and structure–function analysis demonstrated that the polyglutamine region augmented full-length huntingtin PRC2 stimulation, both in \(Hdh^{Q111}\) EBs and in vitro, with reconstituted PRC2. Knowledge of full-length huntingtin's α-helical organization and role as a facilitator of the multi-subunit PRC2 complex provides a novel starting point for studying PRC2 regulation, implicates this chromatin repressive complex in a neurodegenerative disorder and sets the stage for further study of huntingtin's molecular function and the impact of its modulatory polyglutamine region.
- Published
- 2009
- Full Text
- View/download PDF
128. A novel approach to investigate tissue-specific trinucleotide repeat instability
- Author
-
Walker, John R, Wheeler, Vanessa C, Gusella, James F, MacDonald, Marcy E, Lee, Jong-Min, Kang, Kihwa, Su, Andrew I, Gillis, Tammy, Dragileva, Ella, Boily, Marie-Josee, Zhang, Jie, Kohane, Isaac, Wiltshire, Tim, Cyr, Michel, and Lopez, Edith T
- Subjects
3. Good health - Abstract
Background In Huntington's disease (HD), an expanded CAG repeat produces characteristic striatal neurodegeneration. Interestingly, the HD CAG repeat, whose length determines age at onset, undergoes tissue-specific somatic instability, predominant in the striatum, suggesting that tissue-specific CAG length changes could modify the disease process. Therefore, understanding the mechanisms underlying the tissue specificity of somatic instability may provide novel routes to therapies. However progress in this area has been hampered by the lack of sensitive high-throughput instability quantification methods and global approaches to identify the underlying factors. Results Here we describe a novel approach to gain insight into the factors responsible for the tissue specificity of somatic instability. Using accurate genetic knock-in mouse models of HD, we developed a reliable, high-throughput method to quantify tissue HD CAG repeat instability and integrated this with genome-wide bioinformatic approaches. Using tissue instability quantified in 16 tissues as a phenotype and tissue microarray gene expression as a predictor, we built a mathematical model and identified a gene expression signature that accurately predicted tissue instability. Using the predictive ability of this signature we found that somatic instability was not a consequence of pathogenesis. In support of this, genetic crosses with models of accelerated neuropathology failed to induce somatic instability. In addition, we searched for genes and pathways that correlated with tissue instability. We found that expression levels of DNA repair genes did not explain the tissue specificity of somatic instability. Instead, our data implicate other pathways, particularly cell cycle, metabolism and neurotransmitter pathways, acting in combination to generate tissue-specific patterns of instability. Conclusion Our study clearly demonstrates that multiple tissue factors reflect the level of somatic instability in different tissues. In addition, our quantitative, genome-wide approach is readily applicable to high-throughput assays and opens the door to widespread applications with the potential to accelerate the discovery of drugs that alter tissue instability.
129. Population-specific genetic modification of Huntington's disease in Venezuela
- Author
-
Chao, Michael J., Kim, Kyung-Hee, Shin, Jun Wan, Lucente, Diane, Wheeler, Vanessa, Li, Hong, Roach, Jared C., Hood, Leroy, Wexler, Nancy S., Jardim, Laura B., Holmans, Peter, Jones, Lesley, Orth, Michael, Kwak, Seung, MacDonald, Marcy, Gusella, James, and Lee, Jong Min
- Subjects
FOS: Biological sciences ,Therapeutics--Research ,FOS: Clinical medicine ,Genetics ,Neurosciences ,Huntington's disease ,Neurogenetics ,3. Good health - 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.
130. Corrigendum: Motivational, proteostatic and transcriptional deficits precede synapse loss, gliosis and neurodegeneration in the B6.HttQ111/+ model of Huntington's disease.
- Author
-
Bragg, Robert M., Coffey, Sydney R., Weston, Rory M., Ament, Seth A., Cantle, Jeffrey P., Minnig, Shawn, Funk, Cory C., Shuttleworth, Dominic D., Woods, Emily L., Sullivan, Bonnie R., Jones, Lindsey, Glickenhaus, Anne, Anderson, John S., Anderson, Michael D., Dunnett, Stephen B., Wheeler, Vanessa C., MacDonald, Marcy E., Brooks, Simon P., Price, Nathan D., and Carroll, Jeffrey B.
- Published
- 2017
- Full Text
- View/download PDF
131. Long glutamine tracts cause nuclear localization of a novel form of huntingtin in medium spiny striatal neurons in Hdh [sup Q92] and Hdh [sup Q111] knock-in mice.
- Author
-
Wheeler, Vanessa C. and White, Jacqueline K.
- Abstract
Focuses on Huntington's disease (HD) which is caused by an expanded N-terminal glutamine tract that endows huntingtin with a striatal-selective structural property ultimately toxic to medium spiny neurons. Changes in huntingtin's physical properties in precise genetic models of juvenile HD, Hdh[sup Q92] and Hdh[sup Q111] knock-in mice; Glutamine length dependence and dominant inheritance with recruitment of wild-type protein.
- Published
- 2000
- Full Text
- View/download PDF
132. The HD Mutation Does Not Alter Neuronal Death in the Striatum of HdhQ92 Knock-in Mice after Mild Focal Ischemia
- Author
-
Namura, Shobu, Hirt, Lorenz, Wheeler, Vanessa C., McGinnis, Kim M., Hilditch-Maguire, Paige, Moskowitz, Michael A., MacDonald, Marcy E., and Persichetti, Francesca
- Subjects
- *
HUNTINGTON disease , *GLUTAMINE , *GENETIC mutation - Abstract
Huntington''s disease, with its dominant loss of striatal neurons, is triggered by an expanded glutamine tract in huntingtin. To investigate a proposed role for increased activation of the apoptotic cascade in mutant huntingtin''s trigger mechanism, we examined huntingtin cleavage and lesion severity after mild ischemic injury in HdhQ92 mice. We found activation of calpain and caspase proteases and proteolysis of huntingtin in lesioned striatum. However, huntingtin fragments resembled products of calpain I, not caspase-3, cleavage and turnover was accompanied by augmented levels of full-length normal and mutant protein. By contrast, the number of apoptotic cells, total and striatal infarct size, and degree of neurologic deficit were similar in HdhQ92 and wild-type mice, indicating that the disease process neither strongly protected nor sensitized striatal neurons to apoptotic death. Thus, our findings do not support a role for increased apoptosis or caspase-3 cleavage in the mechanism by which mutant huntingtin triggers disease. However, they suggest that calpain activation and huntingtin regulation merit investigation as modifiers of disease progression in neurons injured by the harmful consequences of full-length mutant huntingtin. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
133. FAN1, a DNA Repair Nuclease, as a Modifier of Repeat Expansion Disorders
- Author
-
Gagan B. Panigrahi, Jean-Yves Masson, Amit Laxmikant Deshmukh, Antonio Porro, Mohiuddin Mohiuddin, Stella Lanni, Christopher E. Pearson, Alessandro A. Sartori, Marie-Christine Caron, University of Zurich, Jones, Lesley, Pearson, Christopher E, and Wheeler, Vanessa
- Subjects
0301 basic medicine ,DNA Repair ,DNA repair ,2804 Cellular and Molecular Neuroscience ,Clinical Neurology ,610 Medicine & health ,Review ,Biology ,Genomic Instability ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Huntington's disease ,FAN1 ,medicine ,Animals ,Humans ,Spinocerebellar Ataxias ,Copy-number variation ,nuclease ,Gene ,repeat instability ,Genetics ,modifier ,Endodeoxyribonucleases ,Genes, Modifier ,10061 Institute of Molecular Cancer Research ,medicine.disease ,Multifunctional Enzymes ,FMR1 ,karyomegalic interstitial nephritis ,Exodeoxyribonucleases ,Huntington Disease ,2728 Neurology (clinical) ,030104 developmental biology ,Spinocerebellar ataxia ,570 Life sciences ,biology ,Neurology (clinical) ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Huntington’s disease - Abstract
FAN1 encodes a DNA repair nuclease. Genetic deficiencies, copy number variants, and single nucleotide variants of FAN1 have been linked to karyomegalic interstitial nephritis, 15q13.3 microdeletion/microduplication syndrome (autism, schizophrenia, and epilepsy), cancer, and most recently repeat expansion diseases. For seven CAG repeat expansion diseases (Huntington’s disease (HD) and certain spinocerebellar ataxias), modification of age of onset is linked to variants of specific DNA repair proteins. FAN1 variants are the strongest modifiers. Non-coding disease-delaying FAN1 variants and coding disease-hastening variants (p.R507H and p.R377W) are known, where the former may lead to increased FAN1 levels and the latter have unknown effects upon FAN1 functions. Current thoughts are that ongoing repeat expansions in disease-vulnerable tissues, as individuals age, promote disease onset. Fan1 is required to suppress against high levels of ongoing somatic CAG and CGG repeat expansions in tissues of HD and FMR1 transgenic mice respectively, in addition to participating in DNA interstrand crosslink repair. FAN1 is also a modifier of autism, schizophrenia, and epilepsy. Coupled with the association of these diseases with repeat expansions, this suggests a common mechanism, by which FAN1 modifies repeat diseases. Yet how any of the FAN1 variants modify disease is unknown. Here, we review FAN1 variants, associated clinical effects, protein structure, and the enzyme’s attributed functional roles. We highlight how variants may alter its activities in DNA damage response and/or repeat instability. A thorough awareness of the FAN1 gene and FAN1 protein functions will reveal if and how it may be targeted for clinical benefit.
- Published
- 2021
- Full Text
- View/download PDF
134. Suppression of Huntington's Disease Somatic Instability by Transcriptional Repression and Direct CAG Repeat Binding.
- Author
-
Mathews EW, Coffey SR, Gärtner A, Belgrad J, Bragg RM, O'Reilly D, Cantle JP, McHugh C, Summers A, Fentz J, Schwagarus T, Cornelius A, Lingos I, Burch Z, Kovalenko M, Andrew MA, Frank Bennett C, Kordasiewicz HB, Marchionini DM, Wilkinson H, Vogt TF, Pinto RM, Khvorova A, Howland D, Wheeler VC, and Carroll JB
- Abstract
Huntington's disease (HD) arises from a CAG expansion in the huntingtin ( HTT ) gene beyond a critical threshold. A major thrust of current HD therapeutic development is lowering levels of mutant HTT mRNA (m HTT ) and protein (mHTT) with the aim of reducing the toxicity of these product(s). Human genetic data also support a key role for somatic instability (SI) in HTT 's CAG repeat - whereby it lengthens with age in specific somatic cell types - as a key driver of age of motor dysfunction onset. Thus, an attractive HD therapy would address both mHTT toxicity and SI, but to date the relationship between SI and HTT lowering remains unexplored. Here, we investigated multiple therapeutically-relevant HTT-lowering modalities to establish the relationship between HTT lowering and SI in HD knock-in mice. We find that repressing transcription of mutant Htt (m Htt ) provides robust protection from SI, using diverse genetic and pharmacological approaches (antisense oligonucleotides, CRISPR-Cas9 genome editing, the Lac repressor, and virally delivered zinc finger transcriptional repressor proteins, ZFPs). However, we find that small interfering RNA (siRNA), a potent HTT-lowering treatment, lowers HTT levels without influencing SI and that SI is also normal in mice lacking 50% of total HTT levels, suggesting HTT levels, per se , do not modulate SI in trans . Remarkably, modified ZFPs that bind the m Htt locus, but lack a repressive domain, robustly protect from SI, despite not reducing HTT mRNA or protein levels. These results have important therapeutic implications in HD, as they suggest that DNA-targeted HTT-lowering treatments may have significant advantages compared to other HTT-lowering approaches, and that interaction of a DNA-binding protein and HTT' s CAG repeats may provide protection from SI while sparing HTT expression., Competing Interests: Declaration of Interests A.N., A.S., J.F., T.S., A.C., I.L. are Employees of Evotec, and may have stock options. C.F.B. and H.K. Are full time employees at, and hold shares in, Ionis Pharmaceuticals. V.C.W. was a founding scientific advisory board member with a financial interest in Triplet Therapeutics Inc., her financial interests were reviewed and are managed by Massachusetts General Hospital (MGH) and Mass General Brigham (MGB) in accordance with their conflict of interest policies. V.C.W. is a scientific advisory board member of LoQus23 Therapeutics Ltd. and has provided paid consulting services to Acadia Pharmaceuticals Inc., Alnylam Inc., Biogen Inc., Passage Bio and Rgenta Therapeutics and has received research support from Pfizer Inc. J.B.C. Has provided paid consulting and/or conducted sponsored research for Wave Life Sciences, Skyhawk Therapeutics, Cajal Neuroscience, Ionis Pharmaceuticals, and Alnylam, and Gudiepoint. D.H., D.M. and T.V.are full-time employees of CHDI Foundation. A.K. discloses ownership of stocks in RXi Pharmaceuticals and Advirna, and is a founder of Atalanta Therapeutics and Comanche Biopharma. R.B. received consulting fees from Takeda.
- Published
- 2024
- Full Text
- View/download PDF
135. Genetic modifiers of somatic expansion and clinical phenotypes in Huntington's disease reveal shared and tissue-specific effects.
- Author
-
Lee JM, McLean ZL, Correia K, Shin JW, Lee S, Jang JH, Lee Y, Kim KH, Choi DE, Long JD, Lucente D, Seong IS, Pinto RM, Giordano JV, Mysore JS, Siciliano J, Elezi E, Ruliera J, Gillis T, Wheeler VC, MacDonald ME, Gusella JF, Gatseva A, Ciosi M, Lomeikaite V, Loay H, Monckton DG, Wills C, Massey TH, Jones L, Holmans P, Kwak S, Sampaio C, Orth M, Bernhard Landwehrmeyer G, Paulsen JS, Ray Dorsey E, and Myers RH
- Abstract
Huntington's disease (HD), due to expansion of a CAG repeat in HTT , is representative of a growing number of disorders involving somatically unstable short tandem repeats. We find that overlapping and distinct genetic modifiers of clinical landmarks and somatic expansion in blood DNA reveal an underlying complexity and cell-type specificity to the mismatch repair-related processes that influence disease timing. Differential capture of non-DNA-repair gene modifiers by multiple measures of cognitive and motor dysfunction argues additionally for cell-type specificity of pathogenic processes. Beyond trans modifiers, differential effects are also illustrated at HTT by a 5'-UTR variant that promotes somatic expansion in blood without influencing clinical HD, while, even after correcting for uninterrupted CAG length, a synonymous sequence change at the end of the CAG repeat dramatically hastens onset of motor signs without increasing somatic expansion. Our findings are directly relevant to therapeutic suppression of somatic expansion in HD and related disorders and provide a route to define the individual neuronal cell types that contribute to different HD clinical phenotypes.
- Published
- 2024
- Full Text
- View/download PDF
136. Base editing strategies to convert CAG to CAA diminish the disease-causing mutation in Huntington's disease.
- Author
-
Choi DE, Shin JW, Zeng S, Hong EP, Jang JH, Loupe JM, Wheeler VC, Stutzman HE, Kleinstiver B, and Lee JM
- Subjects
- Animals, Mice, Disease Models, Animal, Humans, Mutation, Gene Knock-In Techniques, Huntington Disease genetics, Huntington Disease therapy, Gene Editing methods, Huntingtin Protein genetics, Huntingtin Protein metabolism, Trinucleotide Repeat Expansion genetics
- Abstract
An expanded CAG repeat in the huntingtin gene ( HTT ) causes Huntington's disease (HD). Since the length of uninterrupted CAG repeat, not polyglutamine, determines the age-at-onset in HD, base editing strategies to convert CAG to CAA are anticipated to delay onset by shortening the uninterrupted CAG repeat. Here, we developed base editing strategies to convert CAG in the repeat to CAA and determined their molecular outcomes and effects on relevant disease phenotypes. Base editing strategies employing combinations of cytosine base editors and guide RNAs (gRNAs) efficiently converted CAG to CAA at various sites in the CAG repeat without generating significant indels, off-target edits, or transcriptome alterations, demonstrating their feasibility and specificity. Candidate BE strategies converted CAG to CAA on both expanded and non-expanded CAG repeats without altering HTT mRNA and protein levels. In addition, somatic CAG repeat expansion, which is the major disease driver in HD, was significantly decreased in the liver by a candidate BE strategy treatment in HD knock-in mice carrying canonical CAG repeats. Notably, CAG repeat expansion was abolished entirely in HD knock-in mice carrying CAA-interrupted repeats, supporting the therapeutic potential of CAG-to-CAA conversion strategies in HD and potentially other repeat expansion disorders., Competing Interests: DC, JS, SZ, EH, JJ, JL, HS No competing interests declared, VW V.C.W. was a founding scientific advisory board member with financial interest in Triplet Therapeutics Inc, Her financial interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict of interest policies. V.C.W. is a scientific advisory board member of LoQus23 Therapeutics Ltd. and has provided paid consulting services to Acadia Pharmaceuticals Inc, Alnylam Inc, Biogen Inc and Passage Bio. V.C.W. has received research support from Pfizer Inc, BK B.P.K is an inventor on patents and/or patent applications filed by Mass General Brigham that describe genome engineering technologies. B.P.K. is a consultant for EcoR1 capital and is a scientific advisory board member of Acrigen Biosciences, Life Edit Therapeutics, and Prime Medicine, JL J-ML consults for GenKOre and serves in the advisory board of GenEdit Inc, (© 2023, Choi et al.)
- Published
- 2024
- Full Text
- View/download PDF
137. Identification of genetic modifiers of Huntington's disease somatic CAG repeat instability by in vivo CRISPR-Cas9 genome editing.
- Author
-
Mouro Pinto R, Murtha R, Azevedo A, Douglas C, Kovalenko M, Ulloa J, Crescenti S, Burch Z, Oliver E, Vitalo A, Mota-Silva E, Riggs MJ, Correia K, Elezi E, Demelo B, Carroll JB, Gillis T, Gusella JF, MacDonald ME, and Wheeler VC
- Abstract
Huntington's disease (HD), one of >50 inherited repeat expansion disorders (Depienne and Mandel, 2021), is a dominantly-inherited neurodegenerative disease caused by a CAG expansion in HTT (The Huntington's Disease Collaborative Research Group, 1993). Inherited CAG repeat length is the primary determinant of age of onset, with human genetic studies underscoring that the property driving disease is the CAG length-dependent propensity of the repeat to further expand in brain (Swami et al ., 2009; GeM-HD, 2015; Hensman Moss et al ., 2017; Ciosi et al ., 2019; GeM-HD, 2019; Hong et al ., 2021). Routes to slowing somatic CAG expansion therefore hold great promise for disease-modifying therapies. Several DNA repair genes, notably in the mismatch repair (MMR) pathway, modify somatic expansion in HD mouse models (Wheeler and Dion, 2021). To identify novel modifiers of somatic expansion, we have used CRISPR-Cas9 editing in HD knock-in mice to enable in vivo screening of expansion-modifier candidates at scale. This has included testing of HD onset modifier genes emerging from human genome-wide association studies (GWAS), as well as interactions between modifier genes, thereby providing new insight into pathways underlying CAG expansion and potential therapeutic targets.
- Published
- 2024
- Full Text
- View/download PDF
138. Mapping SCA1 regional vulnerabilities reveals neural and skeletal muscle contributions to disease.
- Author
-
Duvick L, Southern WM, Benzow KA, Burch ZN, Handler HP, Mitchell JS, Kuivinen H, Gadiparthi U, Yang P, Soles A, Sheeler CA, Rainwater O, Serres S, Lind EB, Nichols-Meade T, You Y, O'Callaghan B, Zoghbi HY, Cvetanovic M, Wheeler VC, Ervasti JM, Koob MD, and Orr HT
- Subjects
- Animals, Mice, Humans, Male, Mice, Transgenic, Gene Knock-In Techniques, Female, Phenotype, Neurons metabolism, Neurons pathology, Ataxin-1 genetics, Ataxin-1 metabolism, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias pathology, Muscle, Skeletal pathology, Muscle, Skeletal metabolism, Disease Models, Animal
- Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1) protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockin mouse (f-ATXN1146Q/2Q) with mouse Atxn1 coding exons replaced by human ATXN1 exons encoding 146 glutamines. f-ATXN1146Q/2Q mice manifested SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. Central nervous system (CNS) contributions to disease were revealed using f-ATXN1146Q/2Q;Nestin-Cre mice, which showed improved rotarod, open field, and Barnes maze performance by 6-12 weeks of age. In contrast, striatal contributions to motor deficits using f-ATXN1146Q/2Q;Rgs9-Cre mice revealed that mice lacking ATXN1146Q/2Q in striatal medium-spiny neurons showed a trending improvement in rotarod performance at 30 weeks of age. Surprisingly, a prominent role for muscle contributions to disease was revealed in f-ATXN1146Q/2Q;ACTA1-Cre mice based on their recovery from kyphosis and absence of muscle pathology. Collectively, data from the targeted conditional deletion of the expanded allele demonstrated CNS and peripheral contributions to disease and highlighted the need to consider muscle in addition to the brain for optimal SCA1 therapeutics.
- Published
- 2024
- Full Text
- View/download PDF
139. PMS1 as a target for splice modulation to prevent somatic CAG repeat expansion in Huntington's disease.
- Author
-
McLean ZL, Gao D, Correia K, Roy JCL, Shibata S, Farnum IN, Valdepenas-Mellor Z, Rapuru M, Morini E, Ruliera J, Gillis T, Lucente D, Kleinstiver BP, Lee JM, MacDonald ME, Wheeler VC, Pinto RM, and Gusella JF
- Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder whose motor, cognitive, and behavioral manifestations are caused by an expanded, somatically unstable CAG repeat in the first exon of HTT that lengthens a polyglutamine tract in huntingtin. Genome-wide association studies (GWAS) have revealed DNA repair genes that influence the age-at-onset of HD and implicate somatic CAG repeat expansion as the primary driver of disease timing. To prevent the consequent neuronal damage, small molecule splice modulators (e.g., branaplam) that target HTT to reduce the levels of huntingtin are being investigated as potential HD therapeutics. We found that the effectiveness of the splice modulators can be influenced by genetic variants, both at HTT and other genes where they promote pseudoexon inclusion. Surprisingly, in a novel hTERT-immortalized retinal pigment epithelial cell (RPE1) model for assessing CAG repeat instability, these drugs also reduced the rate of HTT CAG expansion. We determined that the splice modulators also affect the expression of the mismatch repair gene PMS1 , a known modifier of HD age-at-onset. Genome editing at specific HTT and PMS1 sequences using CRISPR-Cas9 nuclease confirmed that branaplam suppresses CAG expansion by promoting the inclusion of a pseudoexon in PMS1 , making splice modulation of PMS1 a potential strategy for delaying HD onset. Comparison with another splice modulator, risdiplam, suggests that other genes affected by these splice modulators also influence CAG instability and might provide additional therapeutic targets., Competing Interests: Competing interests J.F.G. and V.C.W. were founding scientific advisory board members with a financial interest in Triplet Therapeutics Inc. Their financial interests were reviewed and are managed by Massachusetts General Hospital (MGH) and Mass General Brigham (MGB) in accordance with their conflict of interest policies. J.F.G. consults for Transine Therapeutics, Inc. and has previously provided paid consulting services to Wave Therapeutics USA Inc., Biogen Inc. and Pfizer Inc. V.C.W. is a scientific advisory board member of LoQus23 Therapeutics Ltd. and has provided paid consulting services to Acadia Pharmaceuticals Inc., Alnylam Inc., Biogen Inc. and Passage Bio. R.M.P. and V.C.W. have received research support from Pfizer Inc. B.P.K. is a consultant for EcoR1 capital and Curie.Bio, and is an advisor to Acrigen Biosciences, Life Edit Therapeutics and Prime Medicine. B.P.K. has a financial interest in Prime Medicine, Inc., a company developing therapeutic CRISPR-Cas technologies for gene editing. B.P.K.’s interests were reviewed and are managed by MGH and MGB in accordance with their conflict-of-interest policies. J-M.L. consults for Life Edit Therapeutics and serves on the scientific advisory board of GenEdit Inc. E.M. is inventor on an International Patent Application Number PCT/US2021/012103, assigned to Massachusetts General Hospital and PTC Therapeutics entitled “RNA Splicing Modulation” related to use of BPN-15477 in modulating splicing.
- Published
- 2023
- Full Text
- View/download PDF
140. Delineating regional vulnerability in the neurodegenerative disease SCA1 using a conditional mutant ATXN1 mouse.
- Author
-
Duvick L, Southern WM, Benzow K, Burch ZN, Handler HP, Mitchell JS, Kuivinen H, Gadiparthi UK, Yang P, Soles A, Scheeler C, Rainwater O, Serres S, Lind E, Nichols-Meade T, O'Callaghan B, Zoghbi HY, Cvetanovic M, Wheeler VC, Ervasti JM, Koob MD, and Orr HT
- Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ATXN1 protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockout mouse model ( f-ATXN1
146Q/2Q ) having mouse Atxn1 coding exons replaced by human exons encoding 146 glutamines. F-ATXN1146Q/2Q mice manifest SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. CNS contributions to disease were revealed using ATXN1146Q/2Q ; Nestin-Cre mice, that showed improved rotarod, open field and Barnes maze performances. Striatal contributions to motor deficits were examined using f-ATXN1146Q/2Q ; Rgs9-Cre mice. Mice lacking striatal ATXN1146Q/2Q had improved rotarod performance late in disease. Muscle contributions to disease were revealed in f-ATXN1146Q/2Q ; ACTA1-Cre mice which lacked muscle pathology and kyphosis seen in f-ATXN1146Q/2Q mice. Kyphosis was not improved in f-ATXN1146Q/2Q ;Nestin- Cre mice. Thus, optimal SCA1 therapeutics will require targeting mutant ATXN1 toxic actions in multiple brain regions and muscle.- Published
- 2023
- Full Text
- View/download PDF
141. Base editing strategies to convert CAG to CAA diminish the disease-causing mutation in Huntington's disease.
- Author
-
Choi DE, Shin JW, Zeng S, Hong EP, Jang JH, Loupe JM, Wheeler VC, Stutzman HE, Kleinstiver BP, and Lee JM
- Abstract
An expanded CAG repeat in the huntingtin gene ( HTT ) causes Huntington's disease (HD). Since the length of uninterrupted CAG repeat, not polyglutamine, determines the age-at-onset in HD, base editing strategies to convert CAG to CAA are anticipated to delay onset by shortening the uninterrupted CAG repeat. Here, we developed base editing strategies to convert CAG in the repeat to CAA and determined their molecular outcomes and effects on relevant disease phenotypes. Base editing strategies employing combinations of cytosine base editors and gRNAs efficiently converted CAG to CAA at various sites in the CAG repeat without generating significant indels, off-target edits, or transcriptome alterations, demonstrating their feasibility and specificity. Candidate BE strategies converted CAG to CAA on both expanded and non-expanded CAG repeats without altering HTT mRNA and protein levels. In addition, somatic CAG repeat expansion, which is the major disease driver in HD, was significantly decreased by a candidate BE strategy treatment in HD knock-in mice carrying canonical CAG repeats. Notably, CAG repeat expansion was abolished entirely in HD knock-in mice carrying CAA-interrupted repeats, supporting the therapeutic potential of CAG-to-CAA conversion base editing strategies in HD and potentially other repeat expansion disorders.
- Published
- 2023
- Full Text
- View/download PDF
142. Corrigendum: Motivational, proteostatic and transcriptional deficits precede synapse loss, gliosis and neurodegeneration in the B6.Htt Q111/+ model of Huntington's disease.
- Author
-
Bragg RM, Coffey SR, Weston RM, Ament SA, Cantle JP, Minnig S, Funk CC, Shuttleworth DD, Woods EL, Sullivan BR, Jones L, Glickenhaus A, Anderson JS, Anderson MD, Dunnett SB, Wheeler VC, MacDonald ME, Brooks SP, Price ND, and Carroll JB
- Published
- 2017
- Full Text
- View/download PDF
143. Systematic behavioral evaluation of Huntington's disease transgenic and knock-in mouse models.
- Author
-
Menalled L, El-Khodor BF, Patry M, Suárez-Fariñas M, Orenstein SJ, Zahasky B, Leahy C, Wheeler V, Yang XW, MacDonald M, Morton AJ, Bates G, Leeds J, Park L, Howland D, Signer E, Tobin A, and Brunner D
- Subjects
- Aging, Animals, Female, Gene Knock-In Techniques, Genotype, Humans, Huntingtin Protein, Male, Mice, Mice, Transgenic, Nerve Tissue Proteins genetics, Neuropsychological Tests, Nuclear Proteins genetics, Phenotype, Sex Characteristics, Behavior, Animal, Disease Models, Animal, Huntington Disease genetics, Motor Activity genetics
- Abstract
Huntington's disease (HD) is one of the few neurodegenerative diseases with a known genetic cause, knowledge that has enabled the creation of animal models using genetic manipulations that aim to recapitulate HD pathology. The study of behavioral and neuropathological phenotypes of these HD models, however, has been plagued by inconsistent results across laboratories stemming from the lack of standardized husbandry and testing conditions, in addition to the intrinsic differences between the models. We have compared different HD models using standardized conditions to identify the most robust phenotypic differences, best suited for preclinical therapeutic efficacy studies. With a battery of tests of sensory-motor function, such as the open field and prepulse inhibition tests, we replicate previous results showing a strong and progressive behavioral deficit in the R6/2 line with an average of 129 CAG repeats in a mixed CBA/J and C57BL/6J background. We present the first behavioral characterization of a new model, an R6/2 line with an average of 248 CAG repeats in a pure C57BL/6J background, which also showed a progressive and robust phenotype. The BACHD in a FVB/N background showed robust and progressive behavioral phenotype, while the YAC128 full-length model on either an FVB/N or a C57BL/6J background generally showed milder deficits. Finally, the Hdh(Q111) knock-in mouse on a CD1 background showed very mild deficits. This first extensive standardized cross-characterization of several HD animal models under standardized conditions highlights several behavioral outcomes, such as hypoactivity, amenable to standardized preclinical therapeutic drug screening.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.